Medicament delivery device for administration of opioid antagonists including formulations for naloxone

- kaleo, Inc.

Medicament delivery devices for administration of opioid antagonists are described herein. In some embodiments, an apparatus includes a housing, a medicament container disposed within the housing and an energy storage member disposed within the housing. The medicament container is filled with a naloxone composition that includes naloxone or salts thereof, a tonicity-adjusting agent, and a pH adjusting agent, whereby the osmolality of the naloxone composition ranges from about 250-350 mOsm and the pH ranges from about 3-5. The energy storage member is configured to produce a force to deliver the naloxone composition.

Skip to: Description  ·  Claims  ·  References Cited  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. application Ser. No. 13/036,720, entitled “Medicament Delivery Device for Administration of Opioid Antagonists Including Formulations for Naloxone,” filed Feb. 28, 2011, the entirety of which is incorporated herein by reference.

BACKGROUND

The embodiments described herein relate generally to medical device and pharmaceutical compositions, and more particularly to a medicament delivery device for administration of opioid antagonists, including formulations for naloxone.

Naloxone is a medicament that prevents and/or reverses the effects of opioids. Known formulations of naloxone can be used, for example, to treat respiratory depression and other indications that result from opioid toxicity. For example, known formulations for naloxone can be used to reverse and/or mitigate the effects of an overdose of a drug containing opioids, such as, for example, heroin. In such situations, it is desirable to deliver the naloxone formulation quickly and in a manner that will produce a rapid onset of action. Accordingly, known formulations of naloxone are often delivered either intranasally or via injection.

The delivery of naloxone intranasally or via injection, however, often involves completing a series of operations that, if not done properly, can limit the effectiveness of the naloxone formulation. For example, prior to delivering the naloxone, the user must first determine whether the patient's symptoms warrant the delivery of naloxone, and then couple a needle (or an atomizer) to a syringe containing the naloxone formulation. After the device is prepared for delivery, the user then selects the region of the body in which the naloxone is to be delivered, and manually produces a force to deliver the naloxone. In some situations, such as, for example, when the patient is in an ambulance or a hospital setting, the user then inserts an intravenous catheter to administer the naloxone. Additionally, after the delivery of the naloxone formulation, the user must dispose of the device properly (e.g., to prevent needle sticks in instances where the naloxone is injected) and seek further medical attention for the patient. Accordingly, known formulations of naloxone are often delivered by a healthcare provider in a controlled environment (e.g. a hospital, physician's office, clinic or the like). Access to emergency medical facilities and/or trained health care providers, however, is not always available when an individual is suffering from an overdose. Moreover, because naloxone is often administered during an emergency situation, even experienced and/or trained users may be subject to confusion and/or panic, thereby compromising the delivery of the naloxone formulation.

Known devices for delivering naloxone also require that the user manually generate the force and/or pressure required to convey the naloxone from the device into the body. For example, to deliver naloxone using known syringes, the user manually depresses a plunger into the syringe body. The force generated by manually depressing a plunger, however, can be sporadic, thus resulting in undesirable fluctuations in the flow of the naloxone and/or incomplete delivery of the full dose. Such fluctuations and variability can be particularly undesirable when the naloxone is being atomized for intranasal delivery. Moreover, in certain situations, the user may be unable to generate sufficient force to provide the desired flow rate and/or flow characteristics (e.g., for an atomizer) of the naloxone.

Additionally, because naloxone is often delivered by a healthcare provider in a controlled environment, known formulations of naloxone are generally stored under controlled conditions, and for limited periods of time. For example, known naloxone formulations are often formulated to be stored between 20 and 25 degrees Celsius. Accordingly, known naloxone formulations are not compatible for being carried by a patient or a third party (e.g., a relative of friend of the patient) for long periods of time.

Thus, a need exists for improved methods and devices for delivering opioid antagonists, such as, for example, devices that provide for the delivery of naloxone by untrained users. Additionally, a need exists for naloxone formulations that can be exposed to a wide range of environmental conditions for long periods of time.

SUMMARY

Medicament delivery devices for administration of opioid antagonists and chemical compositions used within such devices are described herein. In some embodiments, a naloxone composition can be formulated for use in a delivery device of the types shown and described herein. The naloxone composition includes an effective amount of naloxone i.e., 4,5-epoxy-3,14-dihydroxy-17-(2-propenyl)morphinan-6-one, or a pharmaceutically acceptable salt and/or ester thereof. As used herein, an “effective amount” is an amount sufficient to provide a desired therapeutic effect. In some embodiments, the naloxone composition can include a pH adjusting agent, such as, for example, at least one of hydrochloric acid, citric acid, acetic acid, phosphoric acid, or combinations thereof. In some embodiments, the naloxone composition can include one or more tonicity-adjusting agents, such as, for example, at least one of dextrose, glycerin, mannitol, potassium chloride, sodium chloride, or combinations thereof. Because the naloxone composition may be stored in the medicament container of a delivery device for extended periods of time under varying storage conditions, in some embodiments the naloxone composition can include stabilizers to prevent or inhibit decomposition of the naloxone during storage.

In some embodiments, an apparatus includes a housing, a medicament container disposed within the housing and an energy storage member disposed within the housing. The medicament container is filled with a naloxone composition that includes naloxone or salts thereof, a tonicity-adjusting agent, and a pH adjusting agent, whereby the osmolality of the naloxone composition ranges from about 250-350 mOsm and the pH ranges from about 3-5. The energy storage member is configured to produce a force to deliver the naloxone composition.

In some embodiments, the medicament delivery device can further include an elastomeric member disposed within the medicament container that is configured to be compatible with the naloxone composition. Said another way, in some embodiments, an elastomeric member disposed within the medicament container can be formulated to prevent undesired leaching and/or reaction with the naloxone composition. In some embodiments, the elastomeric member is formulated to include a polymer and a curing agent. The polymer includes at least one of bromobutyl or chlorobutyl, and the curing agent includes at least one of sulfur or metal compounds, e.g., metal oxides such as zinc oxide or magnesium oxide, etc.

In some embodiments, the medicament delivery device can include an electronic circuit system coupled to the housing. The electronic circuit system is configured to produce an output when the electronic circuit system is actuated. The output can be, for example, an audible or visual output related to the naloxone composition (e.g., an indication of the expiration date, the symptoms requirement treatment with naloxone or the like), the use of the medicament delivery device, and/or post-administration procedures (e.g., a prompt to call 911, instructions for the disposal of the device or the like).

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 is a schematic illustration of a medicament delivery device according to an embodiment.

FIG. 2 is a schematic illustration of a medicament delivery device according to an embodiment.

FIG. 3 is a schematic illustration of a medicament delivery device according to an embodiment.

FIGS. 4 and 5 are perspective views of a medical injector according to an embodiment, in a first configuration.

FIG. 6 is a front view of the medical injector illustrated in FIG. 4 with the cover removed.

FIG. 7 is a back view of the medical injector illustrated in FIG. 4 with the cover removed.

FIG. 8 is a front view of a portion of the medical injector illustrated in FIG. 4.

FIG. 9 is a perspective view of a portion of the medical injector illustrated in FIG. 4.

FIG. 10 is a bottom perspective view of a housing of the medical injector illustrated in FIG. 4.

FIG. 11 is a top perspective view of a housing of the medical injector illustrated in FIG. 4.

FIG. 12 is a perspective view of a proximal cap of the medical injector illustrated in FIG. 4.

FIGS. 13 and 14 are a front views of a medicament delivery mechanism of the medical injector illustrated in FIG. 4.

FIG. 15 is a perspective view of a portion of the medical injector illustrated in FIG. 4.

FIG. 16 is an exploded view of a medicament container of the medical injector illustrated in FIG. 4.

FIG. 17 is a front view of a portion of the medical injector illustrated in FIG. 4.

FIG. 18 is a back view of an electronic circuit system of the medical injector illustrated in FIG. 4.

FIG. 19 is a front view of a portion of the electronic circuit system of the medical injector illustrated in FIG. 18.

FIG. 20 is a side view of the electronic circuit system of the medical injector illustrated in FIG. 18.

FIG. 21 is a front view of an electronic circuit system housing of the electronic circuit system illustrated in FIG. 18.

FIG. 22 is a perspective view of the electronic circuit system housing of the electronic circuit system illustrated in FIG. 21.

FIG. 23 is a perspective view of a battery clip of the electronic circuit system illustrated in FIG. 18.

FIG. 24 is a perspective view of a portion of an electronic circuit system of the medical injector illustrated in FIG. 4, in a first configuration.

FIG. 25 is a front view of the medical injector illustrated in FIG. 4 in a first configuration showing the electronic circuit system.

FIGS. 26-28 are front views of a portion of the electronic circuit system of the medical injector labeled as Region Z in FIG. 25 in a first configuration, a second configuration, and a third configuration, respectively.

FIGS. 29 and 30 are perspective views of a cover of the medical injector illustrated in FIG. 4.

FIG. 31 is a perspective view of a safety lock of the medical injector illustrated in FIG. 4.

FIG. 32 is a front view of the safety lock of the medical injector illustrated in FIG. 31.

FIG. 33 is a bottom view of the safety lock of the medical injector illustrated in FIG. 31.

FIG. 34 is a perspective view of a needle sheath of the safety lock of the medical injector illustrated in FIG. 31.

FIG. 35 is a perspective view of a base of the medical injector illustrated in FIG. 4.

FIG. 36 is a front view of the base of the medical injector illustrated in FIG. 4.

FIG. 37 is a back view of the medical injector illustrated in FIG. 4 in a second configuration.

FIG. 38 is a back view of the medical injector illustrated in FIG. 4 in a third configuration.

FIG. 39 is a back view of the medical injector illustrated in FIG. 4 in a fourth configuration (i.e., the needle insertion configuration).

FIG. 40 is a front view of the medical injector illustrated in FIG. 4 in the fourth configuration (i.e., the needle insertion configuration).

FIG. 41 is a front view of the medical injector illustrated in FIG. 4 in a fifth configuration (i.e., the injection configuration).

FIG. 42 is a front view of the medical injector illustrated in FIG. 4 in a sixth configuration (i.e., the retraction configuration).

FIG. 43 is a perspective view of a housing of a medical injector according to an embodiment.

FIG. 44 is a perspective view of an electronic circuit system of a medical injector according to an embodiment.

FIG. 45 is a back view of a printed circuit board of the electronic circuit system shown in FIG. 44.

FIG. 46 is a schematic illustration of the electronic circuit system shown in FIG. 44.

FIG. 47 is a perspective cross-sectional view of the housing and the electronic circuit system illustrated in FIG. 43 and FIG. 44 respectively.

FIG. 48 is a cross-sectional perspective view of a portion of the electronic circuit system illustrated in FIG. 36, taken along line X-X in FIG. 47.

FIG. 49 is a schematic illustration of a medicament delivery device according to an embodiment.

FIG. 50 is a schematic illustration of a kit including a medicament container according to an embodiment.

DETAILED DESCRIPTION

Medicament delivery devices for administration of opioid antagonists and chemical compositions used within such devices are described herein. In some embodiments, a naloxone composition can be formulated for use in a delivery device of the types shown and described herein. The naloxone composition includes an effective amount of naloxone i.e., 4,5-epoxy-3,14-dihydroxy-17-(2-propenyl)morphinan-6-one, or a pharmaceutically acceptable salt and/or ester thereof. As used herein, an “effective amount” is an amount sufficient to provide a desired therapeutic effect. In some embodiments, the naloxone composition can include a pH adjusting agent, such as, for example, at least one of hydrochloric acid, citric acid, acetic acid, phosphoric acid, or combinations thereof. In some embodiments, the naloxone composition can include one or more tonicity-adjusting agents, such as, for example, at least one of dextrose, glycerin, mannitol, potassium chloride, sodium chloride, or combinations thereof. Because the naloxone composition may be stored in the medicament container of a delivery device for extended periods of time under varying storage conditions, in some embodiments the naloxone composition can include stabilizers to prevent or inhibit decomposition of the naloxone during storage.

In some embodiments, a medicament delivery device includes a housing, a medicament container disposed within the housing and an energy storage member disposed within the housing. The medicament container is filled with a naloxone composition that includes naloxone or salts thereof, a tonicity-adjusting agent, and a pH adjusting agent, whereby the osmolality of the naloxone composition ranges from about 250-350 mOsm and the pH ranges from about 3-5. The energy storage member is configured to produce a force to deliver the naloxone composition.

In some embodiments, the medicament delivery device can further include an elastomeric member disposed within the medicament container that is configured to be compatible with the naloxone composition. Said another way, in some embodiments, an elastomeric member disposed within the medicament container can be formulated to prevent undesired leaching and/or reaction with the naloxone composition. In some embodiments, the elastomeric member is formulated to include a polymer and a curing agent. The polymer includes at least one of bromobutyl or chlorobutyl, and the curing agent includes at least one of sulfur, zinc or magnesium.

In some embodiments, the medicament delivery device can include an electronic circuit system coupled to the housing. The electronic circuit system is configured to produce an output when the electronic circuit system is actuated. The output can be, for example, an audible or visual output related to the naloxone composition (e.g., an indication of the expiration date, the symptoms requirement treatment with naloxone or the like), the use of the medicament delivery device, and/or post-administration procedures (e.g., a prompt to call 911, instructions for the disposal of the device or the like).

In some embodiments, a medicament delivery device includes a housing, a medicament container disposed within the housing, a delivery member coupled to the medicament container, and an energy storage member. The medicament container is filled with a naloxone composition. The energy storage member is disposed within the housing, and is configured to produce a force to deliver the naloxone composition from the medicament container via the delivery member such that the delivery member atomizes the naloxone composition.

In some embodiments, a kit includes a case and a medicament container movably disposed within the case. The medicament container filled with a naloxone composition. The medicament container includes a delivery member coupled thereto. The delivery member can be, for example, a needle, an atomizer or any other mechanism through which the naloxone composition can be conveyed from the medicament container into a body.

As used in this specification and the appended claims, the words “proximal” and “distal” refer to direction closer to and away from, respectively, an operator of the medical device. Thus, for example, the end of the medicament delivery device contacting the patient's body would be the distal end of the medicament delivery device, while the end opposite the distal end would be the proximal end of the medicament delivery device.

Throughout the present specification, the terms “about” and/or “approximately” may be used in conjunction with numerical values and/or ranges. The term “about” is understood to mean those values near to a recited value. For example, “about 40 [units]” may mean within ±25% of 40 (e.g., from 30 to 50), within ±20%, ±15%, ±10%, ±9%, ±8%, ±7%, ±7%, ±5%, ±4%, ±3%, ±2%, ±1%, less than ±1%, or any other value or range of values therein or therebelow. Furthermore, the phrases “less than about [a value]” or “greater than about [a value]” should be understood in view of the definition of the term “about” provided herein. The terms “about” and “approximately” may be used interchangeably.

Throughout the present specification, numerical ranges are provided for certain quantities. It is to be understood that these ranges comprise all subranges therein. Thus, the range “from 50 to 80” includes all possible ranges therein (e.g., 51-79, 52-78, 53-77, 54-76, 55-75, 70-70, etc.). Furthermore, all values within a given range may be an endpoint for the range encompassed thereby (e.g., the range 50-80 includes the ranges with endpoints such as 55-80, 50-75, etc.).

Throughout the present specification, the words “a” or “an” are understood to mean “one or more” unless explicitly stated otherwise. Further, the words “a” or “an” and the phrase “one or more” may be used interchangeably.

Compositions

In one aspect, the present disclosure relates to compositions comprising naloxone or a pharmaceutically acceptable salt thereof suitable for use in the medicament delivery devices disclosed herein. Accordingly, the present naloxone compositions may be adapted for various administration routes, depending on the apparatus in which such composition(s) are to be employed. For example, in some embodiments, the present compositions may adapted for transmucosal administration as, e.g., a nasal spray, or alternatively as a sublingual or buccal spray. In other embodiments, the present naloxone compositions may be adapted for parenteral administration as, e.g., an injectable solution.

The present compositions generally comprise an effective amount of naloxone, i.e., 4,5-epoxy-3,14-dihydroxy-17-(2-propenyl)morphinan-6-one, or a pharmaceutically acceptable salt and/or ester thereof. As used herein, an “effective amount” is an amount sufficient to provide a desired therapeutic effect. For example, as described herein, the present naloxone compositions may be useful in treating respiratory depression and/or other indications associated with opioid toxicity. Accordingly, an effective amount of naloxone in the present compositions may be an amount sufficient to treat such respiratory depression and/or other indications associated with opioid toxicity. The present naloxone compositions typically have a concentration of 4,5-epoxy-3,14-dihydroxy-17-(2-propenyl)morphinan-6-one (or a salt and/or ester thereof) between about 0.01 mg/mL and about 10 mg/mL (e.g., between about 0.05 mg/mL and about 2 mg/mL, or any other value or range of values therein, including about 0.1 mg/mL, about 0.2 mg/mL, about 0.3 mg/mL, about 0.4 mg/mL, about 0.5 mg/mL, about 0.6 mg/mL, about 0.7 mg/mL, about 0.8 mg/mL, about 0.9 mg/mL, about 1.0 mg/mL, about 1.1 mg/mL, about 1.2 mg/mL, about 1.3 mg/mL, about 1.4 mg/mL, about 1.5 mg/mL, about 1.6 mg/mL, about 1.7 mg/mL, about 1.8 mg/mL, or about 1.9 mg/mL).

In some embodiments, the present naloxone compositions comprise a pH adjusting agent. In some embodiments, the pH adjusting agent includes at least one of hydrochloric acid, citric acid, acetic acid, phosphoric acid, or combinations thereof. The pH adjusting agent may comprise an organic and/or inorganic acid or salt thereof (e.g., alkali metal salts [Li, Na, K, etc.], alkaline earth metal [e.g., Ca, Mg, etc.] salts, ammonium salts, etc.). In other embodiments, the pH adjusting agent includes mixtures of one or more acids and one or more salts thereof, e.g., citric acid and citrate salts, acetic acid and acetate salts, phosphoric acid and phosphate salts, etc. In certain embodiments, the pH adjusting agent is added in an amount sufficient to provide a pH of the present naloxone compositions of from about 3 to about 5 (for example a pH of about 3.0, about 3.1, about 3.2, about 3.3, about 3.4, about 3.5, about 3.6, about 3.7, about 3.8, about 3.9, about 4.0, about 4.1, about 4.2, about 4.3, about 4.4, about 4.5, about 4.6, about 4.7, about 4.8, about 4.9, or about 5.0). Accordingly, the present compositions may comprise naloxone salts of the pH adjusting agent employed. For example, in one embodiment, the pH adjusting agent is dilute aqueous hydrochloric acid, and the naloxone salt is naloxone.HCl (e.g., 4,5-epoxy-3,14-dihydroxy-17-(2-propenyl)-morphinan-6-one hydrochloride).

Solvents suitable for use in the present compositions are not particularly limited, provided they are pharmaceutically acceptable. Accordingly, any pharmaceutically acceptable solvent in which the components of the present compositions are soluble, and which does not adversely affect the stability of the present compositions and/or the naloxone and/or naloxone salts contained therein may be employed. For example, in a typical composition, the solvent is sterile water (e.g., USP grade water for injection [WFI]).

In some embodiments, the present compositions may also comprise one or more tonicity-adjusting agents. For example, the tonicity-adjusting agent may include at least one of dextrose, glycerin, mannitol, potassium chloride, sodium chloride, or combinations thereof. The tonicity-adjusting agent(s) may be present in an amount of from about 0.1 mg/mL to about 50 mg/mL (e.g., including about 0.5 mg/mL, about 1.0 mg/mL, about 2.0 mg/mL, about 3.0 mg/mL, about 4.0 mg/mL, about 5.0 mg/mL, about 10 mg/mL, about 15 mg/mL, about 20 mg/mL, about 25 mg/mL, about 30 mg/mL, about 35 mg/mL, about 40 mg/mL, or about 45 mg/mL). In one embodiment, the tonicity-adjusting agent is sodium chloride, and the concentration thereof is between about 0.1 mg/mL and about 20 mg/mL. Generally, in naloxone compositions as described herein which are adapted for injection and/or intranasal delivery, tonicity-adjusting agents are added to provide a desired osmolality. In some embodiments, the osmolality of the naloxone compositions described herein is from about 250 to about 350 mOsm.

Because the naloxone compositions disclosed herein may be stored in the medicament container of the devices described herein for extended periods of time under varying storage conditions, in some embodiments the present compositions may further comprise stabilizers to prevent or inhibit decomposition of the naloxone during storage. Various types of pharmaceutically acceptable stabilizers can be used, including antioxidants (e.g. substituted phenols such as BHT, TBHQ, BHA, or propyl gallate; ascorbates such as ascorboyl palmitate, sodium ascorbate, ascorbic acid), complexing agents (e.g., cyclodextrins); or chelating agents such as EDTA (and its salts), D-gluconic acid δ-lactone, sodium or potassium gluconate, sodium triphosphate, and sodium hexametaphosphate.

EXAMPLES

The chemical stability of several exemplary naloxone hydrochloride compositions were evaluated at various pH and temperature conditions. The formulation of six development lots was performed to evaluate pH and order of addition parameters for naloxone hydrochloride. Assay testing was performed on aliquots of bulk formulation solution sampled prior to the filtration process to determine if the filtration process contributed to any API losses.

Exemplary naloxone compositions were prepared according to the formulations set forth in Table 1, below:

TABLE 1 Exemplary Naloxone Formulations. NaCl API API Mix NaCl Mix Volume of Final Initial Order of Added Time Added Time Initial Adjusted Final pH Adjuster Weight Lot WFI (g) Addition (mg) (seconds) (g) (seconds) pH pH pH (mL) (g) 1 400.01 A 554.73 110 4.5000 98 5.52 3.01 2.99 4.1 500.00 2 400.15 B 555.10 86 4.5269 69 5.41 6.51 6.51 0.5 502.14 3 400.13 A 554.95 104 4.5033 58 5.39 4.47 4.47 0.2 502.17 4 400.00 B 554.58 82 4.4999 87 5.37 3.01 3.01 4.0 502.15 5 399.99 A 554.59 85 4.5513 74 5.40 6.49 6.49 0.2 502.16 6 400.02 B 554.81 68 4.5020 70 5.45 4.50 4.49 0.2 502.19 Final Formulation Solution Density = 1.0043 g/mL (Determined during the formulation process for Lot 1) Order of Addition: A = Water, NaCl, naloxone hydrochloride, pH adjuster B = Water, naloxone hydrochloride, NaCl, pH adjuster

There were no noticeable differences between the formulations from lot to lot. The order of addition of the components had no observable impact on the dissolution times for either the API (Naloxone Hydrochloride) or the NaCl. Initial solution pH values indicated no observable differences between the solutions prior to final pH adjustment. The volumes required for the final pH adjustment were also consistent, indicating no significant differences between the lots.

Solutions were filtered after formulation to determine if filtration after formulation impacts overall solution API concentration. Pre-filtration assay values were consistent with the post-filtration (initial) assay results for each lot, as shown in Table 2, below:

TABLE 2 Filtration of Naloxone Formulations. Pre-Filtration Pest-Filtration Naloxone Hydrochloride Naloxone Hydrochloride Lot (mg/mL) (mg/mL) 1 1.02 1.02 2 1.00 1.00 3 1.01 1.00 4 1.02 1.01 5 1.00 0.99 6 1.01 0.99

Because the naloxone compositions described herein may be stored in the medicament container of the devices described herein for extended periods of time under varying storage conditions, initial testing was performed to support a stability study for the development lots of naloxone hydrochloride. Initial appearance, pH and assay results are shown in Table 3, below:

TABLE 3 Initial Appearance, pH and Assay Results Osmo- Rep- lality Assay Lot licate Appearance (mOsm) pH (mg/mL) 1 1 Clear, colorless solution free 295 3.09 1.02 of visible particulate matter 2 Clear, colorless solution free 295 3.09 1.02 of visible particulate matter Mean (n = 2) 295 3.09 1.02 2 1 Clear, colorless solution free 294 6.54 1.00 of visible particulate matter 2 Clear, colorless solution free 295 6.55 1.00 of visible particulate matter Mean (n = 2) 295 6.55 1.00 3 1 Clear, colorless solution free 292 4.92 1.00 of visible particulate matter 2 Clear, colorless solution free 289 4.96 1.00 of visible particulate matter Mean (n = 2) 291 4.94 1.00 4 1 Clear, colorless solution free 294 3.13 1.01 of visible particulate matter 2 Clear, colorless solution free 294 3.14 1.01 of visible particulate matter Mean (n = 2) 294 3.14 1.01 5 1 Clear, colorless solution free 295 6.57 0.99 of visible particulate matter 2 Clear, colorless solution free 295 6.57 0.99 of visible particulate matter Mean (n = 2) 295 6.57 0.99 6 1 Clear, colorless solution free 292 4.95 0.99 of visible particulate matter 2 Clear, colorless solution free 290 4.99 0.99 of visible particulate matter Mean (n = 2) 291 4.97 0.99

The pH analysis of Lots 3 and 7 exhibited increases of 0.4 and 0.5, respectively, in comparison to the pH values obtained during the formulation process. To verify the initial bulk pH, an aliquot of bulk formulation solution for Lot 7 was removed from storage at 5° C. and allowed to equilibrate to room temperature. The determined pH was 4.52, confirming the final pH obtained during the formulation process. Analysis of related substances was performed for each individual sample, as shown in Table 4, below:

TABLE 4 Initial Related Substance Screening Results Mean of Total Unknown Total Related Related (Identified % Related Substances Substances Lot Replicate by RRT) Substance (%) (%) 1 1 NR NR NR NR 2 NR NR NR 2 1 NR NR NR NR 2 0.559 0.05 0.05 3 1 NR NR NR NR 2 NR NR NR 4 1 NR NR NR NR 2 NR NR NR 5 1 0.160 0.11 0.17 0.09 0.559 0.06 2 NR NR NR 6 1 NR NR NR NR 2 NR NR NR NR = Not Reportable (<0.05% Impurity)

In Table 4, % Related Substance=(Related Substance Peak Area/Total Integrated Area)×100. Peaks greater than or equal to 0.05% were reported. Replicates that exhibited levels of related substances that were not reportable were treated as 0.00% for determination of mean total related substances.

One month stability testing was conducted as previously described, with the following additional analyses:

    • pH analysis for all lots at the 25° C./60% RH condition
    • pH analysis for lots 1 and 4 at the 40° C./75% RH condition
    • Assay and Related Substances analysis for lots 1 and 4 at the 25° C./60% RH and 40° C./75% RH conditions

TABLE 5 One Month Stability Results - 70° C./75% RH Assay Lot Replicate Appearance pH (mg/mL) 1 1 Clear, colorless solution free of visible particulate matter 3.28 1.02 2 Clear, colorless solution free of visible particulate matter 3.25 1.01 Mean (n = 2) 3.27 1.02 2 1 Clear, colorless solution free of visible particulate matter 6.05 0.94 2 Clear, colorless solution free of visible particulate matter 6.05 0.94 Mean (n = 2) 6.05 0.94 3 1 Clear, colorless solution free of visible particulate matter 5.32 0.97 2 Clear, colorless solution free of visible particulate matter 5.44 0.97 Mean (n = 2) 5.38 0.97 4 1 Clear, colorless solution free of visible particulate matter 3.28 1.01 2 Clear, colorless solution free of visible particulate matter 3.28 0.99 Mean (n = 2) 3.28 1.00 5 1 Clear, colorless solution free of visible particulate matter 6.06 0.94 2 Clear, colorless solution free of visible particulate matter 6.05 0.93 Mean (n = 2) 6.06 0.93 6 1 Clear, colorless solution free of visible particulate matter 5.41 0.95 2 Clear, colorless solution free of visible particulate matter 5.27 0.96 Mean (n = 2) 5.34 0.95

TABLE 7a One Month Related Substances Results - 70° C./75% RH - Lots 1-3 Mean of Total Unknown Total Related Related (Identified % Related Substances Substances Lot Replicate by RRT) Substance (%) (%) 1 1 0.038 0.11 0.42 0.46 0.404 0.19 0.597 0.12 2 0.038 0.13 0.50 0.404 0.23 0.597 0.14 2 1 0.034 0.08 4.94 4.77 0.038 0.21 0.089 0.08 0.118 0.08 0.136 4.23 0.403 0.11 1.029 0.15 2 0.034 0.07 4.59 0.038 0.17 0.089 0.08 0.136 4.04 0.403 0.10 1.028 0.13 3 1 0.038 0.19 2.79 2.94 0.118 0.06 0.136 2.17 0.403 0.15 0.596 0.06 1.026 0.17 2 0.038 0.19 3.09 0.117 0.05 0.136 2.46 0.403 0.15 0.596 0.05 1.024 0.19

TABLE 7b One Month Related Substances Results - 70° C./75% RH - Lots 4-6 Mean of Total Unknown Total Related Related (Identified % Related Substances Substances Lot Replicate by RRT) Substance (%) (%) 4 1 0.038 0.11 0.44 0.79 0.403 0.20 0.596 0.13 2 0.039 0.22 1.13 0.116 0.09 0.135 0.09 0.403 0.46 0.596 0.28 5 1 0.039 0.18 4.60 4.80 0.089 0.07 0.115 0.05 0.133 4.20 0.403 0.10 2 0.039 0.18 0.089 0.09 0.132 4.64 0.402 0.08 6 1 0.038 0.17 2.85 2.76 0.132 2.55 0.403 0.13 2 0.038 0.17 2.66 0.114 0.06 0.132 2.31 0.402 0.13

TABLE 7 One Month Stability Results - 40° C./75% RH Lot Replicate pH Assay (mg/mL) 1 1 3.14 1.01 2 3.13 1.01 Mean (n = 2) 3.14 1.01 4 1 3.16 1.01 2 3.16 1.00 Mean (n = 2) 3.16 1.00

TABLE 8 One Month Related Substances Results - 40° C./75% RH Mean of Total Unknown Total Related Related (Identified % Related Substances Substances Lot Replicate by RRT) Substance (%) (%) 1 1 NR NR NR NR 2 NR NR NR 4 1 0.592 0.05 0.05 0.22 2 0.040 0.12 0.38 0.115 0.06 0.592 0.19

TABLE 9 One Month Stability Results - 25° C./60% RH Lot Replicate pH Assay (mg/mL) 1 1 3.11 1.01 2 3.16 1.01 Mean (n = 2) 3.14 1.01 2 1 6.33 No analysis performed 2 6.41 Mean (n = 2) 6.37 3 1 5.20 No analysis performed 2 5.21 Mean (n = 2) 5.21 4 1 3.19 1.01 2 3.17 1.01 Mean (n = 2) 3.18 1.01 5 1 6.32 No analysis performed 2 6.40 Mean (n = 2) 6.36 6 1 5.23 No analysis performed 2 5.24 Mean (n = 2) 5.24

TABLE 10 One Month Related Substances Results - 25° C./60% RH Total Mean of Total Unknown Related Related (Identified % Related Substances Substances Lot Replicate by RRT) Substances (%) (%) 1 1 NR NR NR NR 2 NR NR NR 4 1 NR NR NR NR 2 NR NR NR

Three month stability testing was conducted as previously described, including the following measurements:

    • pH analysis for all lots at the 25° C./60% RH condition
    • pH analysis for lots 1 and 4 at the 40° C./75% RH condition
    • Assay and Related Substances analysis for lots 1 and 4 at the 25° C./60% RH and 40° C./75% RH conditions

TABLE 11 Three Month Stability Results - 70° C./75% RH Assay Lot Replicate Appearance pH (mg/mL) 1 1 Clear, colorless solution free of 3.70 1.00 visible particulate matter 2 Clear, colorless solution free of 3.70 1.00 visible particulate matter Mean (n = 2) 3.70 1.00 4 1 Clear, colorless solution free of 3.74 0.96 visible particulate matter 2 Clear, colorless solution free of 3.77 0.94 visible particulate matter Mean (n = 2) 3.76 0.95

TABLE 12a Three Month Related Substances Results - 70° C./75% RH - Lot 1 Mean of Total Unknown Total Related Related (Identified % Related Substances Substances Lot Replicate by RRT) Substance (%) (%) 1 1 0.039 0.36 1.70 1.74 0.096 0.14 0.136 0.05 0.165 0.34 0.364 0.50 0.384 0.06 0.555 0.19 1.112 0.06 2 0.039 0.39 1.79 0.096 0.15 0.136 0.05 0.165 0.41 0.364 0.47 0.384 0.06 0.555 0.18 1.112 0.07

TABLE 12b Three Month Related Substances Results - 70° C./75% RH - Lot 4 Mean of Total Unknown Total Related Related (Identified % Related Substances Substances Lot Replicate by RRI) Substance (%) (%) 4 1 0.039 0.78 3.44 4.34 0.095 0.38 0.112 0.06 0.135 0.13 0.155 0.06 0.164 0.66 0.312 0.07 0.363 0.76 0.383 0.10 0.554 0.29 1.111 0.14 2 0.039 1.16 5.23 0.096 0.58 0.112 0.11 0.135 0.21 0.155 0.07 0.164 0.96 0.312 0.11 0.363 1.19 0.383 0.14 0.553 0.46 1.110 0.24

TABLE 13 Three Month Stability Results - 40° C./75% RH Assay Lot Replicate Appearance pH (mg/mL) 1 1 Clear, colorless solution free of 3.21 1.01 visible particulate matter 2 Clear, colorless solution free of 3.23 1.01 visible particulate matter Mean (n = 2) 3.22 1.01 4 1 Clear, colorless solution free of 3.31 1.01 visible particulate matter 2 Clear, colorless solution free of 3.33 1.01 visible particulate matter Mean (n = 2) 3.32 1.01

TABLE 14 Three Month Related Substances Results - 40° C./75% RH Mean of Total Unknown Total Related Related (Identified % Related Substances Substances Lot Replicate by RRT) Substance (%) (%) 1 1 0.039 0.06 0.23 0.17 0.364 0.06 0.555 0.11 2 0.555 0.10 0.10 4 1 0.039 0.08 0.25 0.20 0.363 0.06 0.554 0.11 2 0.039 0.05 0.14 0.554 0.09

TABLE 15 Three Month Stability Results - 25° C./60% RH Assay Lot Replicate Appearance pH (mg/mL) 1 1 Clear, colorless solution free of 3.18 1.01 visible particulate matter 2 Clear, colorless solution free of 3.18 1.01 visible particulate matter Mean (n = 2) 3.18 1.01 4 1 Clear, colorless solution free of 3.21 1.01 visible particulate matter 2 Clear, colorless solution free of 3.19 1.01 visible particulate matter Mean (n = 2) 3.20 1.01

TABLE 16 Three Month Related Substances Results - 25° C./60% RH Mean of Total Unknown Total Related Related (Identified % Related Substances Substances Lot Replicate by RRT) Substance (%) (%) 1 1 NR NR NR NR 2 NR NR NR 4 1 NR NR NR NR 2 NR NR NR

Medicament Delivery Devices

The naloxone compositions described herein can be included in any suitable medicament delivery device. For example, in some embodiments, a medicament delivery device configured for self-administration (or administration by an untrained user, such a person accompanying the patient) can include any of the naloxone compositions described herein. Such medicament delivery devices can include, for example, an auto-injector, an intranasal delivery device, a pre-filled syringe, an inhaler or the like. In this manner, the medicament delivery device (including the naloxone composition) can be used by the patient (or an untrained user) in any setting (e.g., the patient's home, in a public venue or the like).

In some embodiments, a medicament delivery device can be configured to automatically deliver any of the naloxone compositions described herein. Similarly stated, in some embodiments, a medicament delivery device, after being actuated by the user, can automatically produce (i.e., produce without any further human intervention) a force to deliver the naloxone composition. In this manner, the force with which the naloxone composition is delivered is within a desired range, and is repeatable between different devices, users or the like.

One example of such a medicament delivery device is provided in FIG. 1, which is a schematic illustration of a medicament delivery device 1000 according to an embodiment. The medicament delivery device 1000 includes a housing 1110, a medicament container 1400 and an energy storage member 1570. The medicament container 1400 is disposed within the housing 1110, and contains (i.e., is filled or partially filled with) a naloxone composition 1420. The energy storage member 1570 is disposed within the housing 1110, and is configured to produce a force F1 to deliver the naloxone composition 1420 (e.g., from the medicament container 1400 to a body).

The naloxone composition 1420 can be any of the naloxone compositions described herein. In particular, the naloxone composition 1420 can include an effective amount of naloxone or salts thereof, a tonicity-adjusting agent, and a pH adjusting agent. The naloxone composition 1420 can be formulated such that the osmolality of the naloxone composition 1420 ranges from about 250-350 mOsm and the pH ranges from about 3-5.

In some embodiments, the naloxone composition 1420 can include any suitable concentration of 4,5-epoxy-3,14-dihydroxy-17-(2-propenyl)morphinan-6-one. In some embodiments, for example, the naloxone composition 1420 has a concentration of 4,5-epoxy-3,14-dihydroxy-17-(2-propenyl)morphinan-6-one between approximately 0.01 mg/mL and approximately 10 mg/mL. In other embodiments, the naloxone composition 1420 has a concentration of 4,5-epoxy-3,14-dihydroxy-17-(2-propenyl)morphinan-6-one between approximately 0.05 mg/mL and approximately 2 mg/mL.

The tonicity-adjusting agent can be any of the tonicity-adjusting agents described herein, and can be included within the naloxone composition 1420 in any suitable amount and/or concentration. For example, in some embodiments, the tonicity-adjusting agent includes at least one of dextrose, glycerin, mannitol, potassium chloride or sodium chloride. In other embodiments, the tonicity-adjusting agent includes sodium chloride in an amount such that a concentration of sodium chloride is between approximately 0.1 mg/mL and approximately 20 mg/mL.

The pH adjusting agent can be any of the pH adjusting agents described herein, and can be included within the naloxone composition 1420 in any suitable amount and/or concentration. For example, in some embodiments, the pH adjusting agent includes at least one of hydrochloric acid, citric acid, citrate salts, acetic acid, acetate salts, phosphoric acid or phosphate salts. In other embodiments, the pH adjusting agent includes a dilute hydrochloric acid.

The medicament container 1400 can be any container suitable for storing the naloxone composition 1420. In some embodiments, the medicament container 1400 can be, for example, a pre-filled syringe, a pre-filled cartridge, a vial, an ampule or the like. In other embodiments, the medicament container 1400 can be a container having a flexible wall, such as, for example, a bladder.

The energy storage member 1570 can be any suitable device or mechanism that, when actuated, produces a force F1 to deliver the naloxone composition 1420. Similarly stated, the energy storage member 1570 can be any suitable device or mechanism that produces the force F1 such that the naloxone composition 1420 is conveyed from the medicament container 1400 into a body of a patient. The naloxone composition 1420 can be conveyed into a body via any suitable mechanism, such as, for example, by injection, intranasally, via inhalation or the like. By employing the energy storage member 1570 to produce the force F1, rather than relying on a user to manually produce the delivery force, the naloxone composition 1420 can be delivered into the body at the desired pressure and/or flow rate, and with the desired characteristics. Moreover, this arrangement reduces the likelihood of partial delivery (e.g., that may result if the user is interrupted or otherwise rendered unable to complete the delivery).

In some embodiments, the energy storage member 1570 can be a mechanical energy storage member, such as a spring, a device containing compressed gas, a device containing a vapor pressure-based propellant or the like. In other embodiments, the energy storage member 1570 can be an electrical energy storage member, such as a battery, a capacitor, a magnetic energy storage member or the like. In yet other embodiments, the energy storage member 1570 can be a chemical energy storage member, such as a container containing two substances that, when mixed, react to produce energy.

As shown in FIG. 1, the energy storage member 1570 can be in any position and/or orientation relative to the medicament container 1400. In some embodiments, for example, the energy storage member 1570 can be positioned within the housing 1110 spaced apart from the medicament container 1400. Moreover, in some embodiments, the energy storage member 1570 can be positioned such that a longitudinal axis of the energy storage member 1570 is offset from the medicament container 1400. In other embodiments, the energy storage member 1570 can substantially surround the medicament container 1400.

Moreover, the energy storage member 1570 can be operably coupled to the medicament container 1400 and/or the naloxone composition 1420 therein such that the force F1 delivers the naloxone composition 1420. In some embodiments, for example, the force F1 can be transmitted to the naloxone composition 1420 via a piston or plunger (not shown in FIG. 1). In other embodiments, the force F1 can be transmitted to the naloxone composition 1420 via a hydraulic or pneumatic coupling. In yet other embodiments, the force F1 can be transmitted to the naloxone composition 1420 electrically. In still other embodiments, the force F1 can be transmitted to the naloxone composition 1420 via a combination of any of the above.

In some embodiments, a medicament container can include an elastomeric member, such that the force produced by an energy storage member is transferred to the naloxone composition by the elastomeric member. For example, FIG. 2 is a schematic illustration of a medicament delivery device 2000 according to an embodiment. The medicament delivery device 2000 includes a housing 2110, a medicament container 2400, an elastomeric member 2410 and an energy storage member 2570. The medicament container 2400 is disposed within the housing 2110, and contains (i.e., is filled or partially filled with) a naloxone composition 2420. The naloxone composition 2420 can be any of the naloxone compositions described herein. The energy storage member 2570 is disposed within the housing 2110, and is configured to produce a force F2 to deliver the naloxone composition 2420, as described herein.

The elastomeric member 2410 is disposed within the medicament container 2400 to seal an end portion of the medicament container 2400. The elastomeric member 2410 can be disposed within the medicament container 2400 during the fill process, and can form a substantially fluid-tight seal to prevent leakage of the naloxone composition 2420 from the medicament container 2400. Moreover, the elastomeric member 2410 is operatively coupled to the energy storage member 2570 such that, in use the force F2 acts upon the elastomeric member 2410 to deliver the naloxone composition 2420 from the medicament container 2400.

The elastomeric member 2410 is formulated to be compatible with the naloxone composition 2420. Similarly stated, the elastomeric member 2410 is formulated to minimize any reduction in the efficacy of the naloxone composition 2420 that may result from contact (either direct or indirect) between the elastomeric member 2410 and the naloxone composition 2420. For example, in some embodiments, the elastomeric member 2410 can be formulated to minimize any leaching or out-gassing of compositions that may have an undesired effect on the naloxone composition 2420. In other embodiments, the elastomeric member 2410 can be formulated to maintain its chemical stability, flexibility and/or sealing properties when in contact (either direct or indirect) with naloxone over a long period of time (e.g., for up to six months, one year, two years, five years or longer).

In some embodiments, the elastomeric member 2410 can be formulated to include a polymer and a curing agent. In such embodiments, the polymer can include at least one of bromobutyl or chlorobutyl. In such embodiments, the curing agent can include at least one of sulfur, zinc or magnesium.

In some embodiments, the elastomeric member 2410 can be constructed from multiple different materials. For example, in some embodiments, at least a portion of the elastomeric member 2410 can be coated. Such coatings can include, for example, polydimethylsiloxane. In some embodiments, at least a portion of the elastomeric member 2410 can be coated with polydimethylsiloxane in an amount of between approximately 0.02 mg/cm2 and approximately 0.80 mg/cm2.

A medicament delivery device configured for delivery of a naloxone composition can include an electronic circuit system that produces an output. Such output can include, for example, any output to assist the user and/or patient in administering the dose of the naloxone composition. For example, FIG. 3 is a schematic illustration of a medicament delivery device 3000 according to an embodiment. The medicament delivery device 3000 includes a housing 3110, a medicament container 3400, an elastomeric member 3410, an energy storage member 3570 and an electronic circuit system 3900. The medicament container 3400 is disposed within the housing 3110, and contains (i.e., is filled or partially filled with) a naloxone composition 3420. The naloxone composition 3420 can be any of the naloxone compositions described herein. For example, in some embodiments, the naloxone composition 3420 can include an effective amount of naloxone or salts thereof, a tonicity-adjusting agent, and a pH adjusting agent. The naloxone composition can be formulated such that the osmolality of the naloxone composition ranges from about 250-350 mOsm and the pH ranges from about 3-5.

The energy storage member 3570 is disposed within the housing 3110, and is configured to produce a force F2 to deliver the naloxone composition 3420, as described herein. The elastomeric member 3410 is disposed within the medicament container 3400 to seal an end portion of the medicament container 3400. Moreover, the elastomeric member 3410 is operatively coupled to the energy storage member 3570 such that, in use the force F2 acts upon the elastomeric member 3410 to deliver the naloxone composition 3420 from the medicament container 3400.

The electronic circuit system 3900 is configured to produce an output OP1 when the electronic circuit system 3900 is actuated. The output can be, for example, an audible or visual output related to the naloxone composition (e.g., an indication of the expiration date, the symptoms requirement treatment with naloxone or the like), the use of the medicament delivery device, and/or post-administration procedures (e.g., a prompt to call 911, instructions for the disposal of the device or the like).

For example, in some embodiments, the electronic output OP1 can be associated with an instruction for using the medicament delivery device 3000. In other embodiments, the electronic output OP1 can be a post-use instruction, such as, for example, a recorded message notifying the user that the delivery of the naloxone composition 3420 is complete, instructing the user on post-use disposal of the medicament delivery device 3000 (e.g., post-use safety procedures), instructing the user to seek post-use medical treatment, and/or the like. In yet other embodiments, the electronic output OP1 can be associated with the patient's compliance in using medicament delivery device 3000.

The electronic output OP1 can be, for example, a visual output such as, for example, a text message to display on a screen (not shown), and/or an LED. In some embodiments, the electronic output OP1 can be an audio output, such as, for example, recorded speech, a series of tones, and/or the like. In other embodiments, the electronic output OP1 can be a wireless signal configured to be received by a remote device.

As described in more detail herein, the electronic circuit system 3900 can include any suitable electronic components operatively coupled to produce and/or output the electronic output OP1 and/or to perform the functions described herein. The electronic circuit system 3900 can be similar to the electronic circuit systems described in U.S. Pat. No. 7,731,686, entitled “Devices, Systems and Methods for Medicament Delivery,” filed Jan. 9, 2007, which is incorporated herein by reference in its entirety.

The electronic circuit system 3900 can be actuated to produce the electronic output OP1 in any suitable manner. For example, in some embodiments, the electronic circuit system 3900 can be associated with an actuation of the medicament delivery device 3000. Said another way, the electronic circuit system 3900 can be configured to output the electronic output OP1 in response to actuation of the medicament delivery device 3000. In other embodiments, the electronic circuit system 3900 can be actuated manually by a switch (not shown in FIG. 3). Such a switch can be actuated (i.e., to actuated the electronic circuit system 3900) by a push button, by removing the medicament delivery device 3000 from a case or cover (not shown in FIG. 3), by receiving a signal from a remote electronic device, and/or any other suitable mechanism. In yet other embodiments, the electronic circuit system 3900 can be actuated by receiving input from the user via a voice prompt system.

The electronic circuit system 3900 can be coupled to and/or disposed within the housing 3110 in any suitable arrangement. For example, in some embodiments, the electronic circuit system 3900 can be coupled to an exterior or outer surface of the housing 3110. In other embodiments, at least a portion of the electronic circuit system 3900 can be disposed within the housing 3110. Moreover, in some embodiments, a portion of the electronic circuit system 1900 is disposed within the housing 3110 such that the portion of the electronic circuit system 3900 is fluidically and/or physically isolated from the medicament container 3400.

The medicament delivery device 3000 can be any suitable device for automatically delivering any of the naloxone compositions described herein. In some embodiments, the medicament delivery device can be a medical injector configured to automatically deliver a naloxone composition. For example, FIGS. 4-42 show a medical injector 4000, according to an embodiment. FIGS. 4-5 are perspective views of the medical injector 4000 in a first configuration (i.e., prior to use). The medical injector 4000 includes a housing 4110, a delivery mechanism 4500 (see e.g., FIGS. 13-14), a medicament container 4400 containing a naloxone composition 4420 (see e.g., FIG. 16), an electronic circuit system 4900 (see e.g., FIGS. 18-28), a cover 4200 (see e.g., FIGS. 29-30), a safety lock 4700 (see e.g., FIGS. 31-34) and a system actuator assembly 4300 (see e.g., FIGS. 13, 15, 35 and 36). A discussion of the components of the medical injector 4000 will be followed by a discussion of the operation of the medical injector 4000.

As shown in FIGS. 6-12, the housing 4110 has a proximal end portion 4140 and a distal end portion 4120. The housing 4110 defines a first status indicator aperture 4150 and a second status indicator aperture 4151. The first status indicator aperture 4150 defined by the housing 4110 is located on a first side of the housing 4110, and the second status indicator aperture 4151 of the housing 4110 is located on a second side of the housing 4110. The status indicator apertures 4150, 4151 can allow a patient to monitor the status and/or contents of the medicament container 4400 contained within the housing 4110. For example, by visually inspecting the status indicator apertures 4150, 4151, a patient can determine whether the medicament container 4400 contains a medicament and/or whether a medicament has been dispensed.

As shown in FIGS. 10 and 11, the housing 4110 defines a gas cavity 4154, a medicament cavity 4157 and an electronic circuit system cavity 4153. The gas cavity 4154 has a proximal end portion 4155 and a distal end portion 4156. The gas cavity 4154 is configured to receive the gas container 4570 and a portion of the system actuator assembly 4300 (e.g., the release member 4340 and the spring 4370, as shown in FIGS. 13 and 14) as described in further detail herein. The proximal end portion 4155 of the gas cavity 4154 is configured to receive the gas container retention member 4580 of the proximal cap 4112 of the housing 4110, as described in further detail herein. The gas cavity 4154 is in fluid communication with the medicament cavity 4157 via a gas passageway 4144 (see e.g., FIG. 12), as described in further detail herein, and the gas cavity 4154 is in fluid communication with a region outside the housing 4110 via a safety lock aperture 4128 (see e.g., FIGS. 10 and 11).

The medicament cavity 4157 is configured to receive the medicament container 4400 and a portion of the delivery mechanism 4500. In particular, the carrier 4520 and the moveable member 4530 of the medicament delivery mechanism 4500 are movably disposed in the medicament cavity 4157. The medicament cavity 4157 is in fluid communication with a region outside the housing 4110 via a needle aperture 4122 (see e.g., FIGS. 10 and 11).

The electronic circuit system cavity 4153 is configured to receive the electronic circuit system 4900. The housing 4110 has protrusions 4149 (see e.g., FIG. 9) configured to stabilize the electronic circuit system 4900 when the electronic circuit system 4900 is disposed within the electronic circuit system cavity 4153. The housing 4110 also defines connection apertures 4152 configured to receive connection protrusions 4171 of the electronic circuit system 4900, and aperture 4145 (see e.g., FIG. 7) configured to receive a portion of a protrusion 4174 of the electronic circuit system 4900. In this manner, the electronic circuit system 4900 can be coupled to the housing 4110 within the electronic circuit system cavity 4153. In other embodiments, the electronic circuit system 4900 can be coupled within the electronic circuit system cavity 4153 by other suitable means such as an adhesive, a clip, a label and/or the like.

The electronic circuit system cavity 4153 is fluidically and/or physically isolated from the gas cavity 4154 and/or the medicament cavity 4157 by a sidewall 4148. The sidewall 4148 can be any suitable structure to isolate the electronic circuit system cavity 4153 within the housing 4110 from the gas cavity 4154 and/or the medicament cavity 4157 within the housing 4110. Similarly, the gas cavity 4154 and the medicament cavity 4157 are separated by a sidewall 4146. In some embodiments, sidewall 4146 can be similar to the sidewall 4148, which isolates the gas cavity 4154 and the medicament cavity 4157 from the electronic circuit system cavity 4153. In other embodiments the gas cavity 4154 can be fluidically and/or physically isolated from the medicament cavity 4157.

The proximal end portion 4140 of the housing 4110 includes a proximal cap 4112, a speaker protrusion 4147 (see e.g., FIGS. 9 and 10), and cover retention protrusions 4142 (see e.g., FIGS. 5 and 7). The speaker protrusion 4147 is configured to maintain a position of an audio output device 4956 of the electronic circuit system 4900 relative to the housing 4110 when the electronic circuit system 4900 is attached to the housing 4110, as described herein. Cover retention protrusions 4142 are configured to be received within corresponding openings 4215 on the cover 4200. In this manner, as described in more detail herein, the cover 4200 can be removably coupled to and disposed about at least a portion of the housing 4110.

As shown in FIG. 12, the proximal cap 4112 includes a gas container retention member 4580 and defines a gas passageway 4144. The gas container retention member 4580 is configured to receive and/or retain a gas container 4570 that can contain a pressurized gas. The gas passageway 4144 is configured to allow for the passage of gas contained in the gas container 4570 from the gas cavity 4154 to the medicament cavity 4157, as further described herein. Said another way, the gas passageway 4144 places the gas cavity 4154 in fluid communication with the medicament cavity 4157.

As shown in FIGS. 8 and 10, the distal end portion 4120 of the housing 4110 defines a battery isolation protrusion aperture 4121, a needle aperture 4122, a safety lock actuator groove 4123, a safety lock aperture 4128, a base actuator groove 4124, base retention recesses 4125A, 4125B, and base rail grooves 4127. The battery isolation protrusion aperture 4121 is configured to receive the battery isolation protrusion 4235 of the cover 4200 (see e.g., FIG. 30), as described in further detail herein.

The needle aperture 4122 is configured to allow the needle 4452 (see e.g., FIGS. 13, 39 and 40) to exit the housing 4110 when the medical injector 4000 is actuated. The portion of the sidewall of the housing 4110 that defines the needle aperture 4122 includes multiple sheath retention protrusions 4126. In some embodiments, the sheath retention protrusions can interact with the a plurality of ribs 4728 of the needle sheath 4720 (see e.g. FIG. 34) to maintain a position of the needle sheath 4720 relative to the safety lock 4700 when the safety lock 4700 is coupled to the housing 4110 and/or when the safety lock 4700 is being removed from the housing 4110.

The safety lock actuator groove 4123 is configured to receive an actuator 4744 of the safety lock 4700. As described in more detail herein, the actuator 4744 is configured to engage and/or activate the electronic circuit system 4900 when the safety lock 4700 is moved with respect to the housing 4110. The safety lock aperture 4128 is configured to receive a safety lock protrusion 4742 (see e.g., FIGS. 30 and 31). As described in more detail below, when the medical injector is in the first configuration (i.e., when the safety lock 4700 is in place prior to use), the safety lock protrusion 4742 is disposed within an opening 4346 between extensions 4343 of a release member 4340 (see e.g., FIGS. 14 and 15) such that activation of the medical injector 4000 is prevented. The safety lock 4700, its components and functions are further described herein.

The distal base retention recesses 4125A are configured to receive the base connection knobs 4358 of the actuator 4302 (also referred to herein as “base 4302,” see e.g., FIG. 35) when the base 4302 is in a first position relative to the housing 4110. The proximal base retention recesses 4125B are configured to receive the base connection knobs 4358 of the base 4302 when the base 4302 is in a second position relative to the housing 4110. The base retention recesses 4125A, 4125B have a tapered proximal sidewall and a non-tapered distal sidewall. This allows the base retention recesses 4125A, 4125B to receive the base connection knobs 4358 such that the base 4302 can move proximally relative to the housing 4110, but cannot move distally relative to the housing 4110. Said another way, the distal base retention recesses 4125A are configured to prevent the base 4302 from moving distally when the base 4302 is in a first position and the proximal base retention recesses 4125B are configured to prevent the base 4302 from moving distally when the base 4302 is in a second position. Similarly stated, the proximal base retention recesses 4125B and the base connection knobs 4358 cooperatively prevent “kickback” after the medical injector 4000 is actuated.

The base actuator groove 4124 is configured to receive an actuator 4311 of the base 4302. As described in more detail herein, the actuator 4311 of the base 4302 is configured to engage the electronic circuit system 4900 when the base 4302 is moved with respect to the housing 4110. The base rail grooves 4127 are configured to receive the guide members 4312 of the base 4302. The guide members 4312 of the base 4302 and the base rail grooves 4127 of the housing 4110 engage each other in a way that allows the guide members 4312 of the base 4302 to slide in a proximal and/or distal direction within the base rail grooves 4127 while limiting lateral movement of the guide members 4312. This arrangement allows the base 4302 to move in a proximal and/or distal direction with respect to the housing 4110 but prevents the base 4302 from moving in a lateral direction with respect to the housing 4110.

FIGS. 13-16 show the medicament container 4400, the system actuator assembly 4300 and the medicament delivery mechanism 4500 of the medical injector 4000. The medical injector 4000 is similar to the auto-injectors described in U.S. Pat. No. 7,648,482, entitled “Devices, Systems and Methods for Medicament Delivery,” filed Nov. 21, 2006, which is incorporated herein by reference in its entirety.

The medicament container 4400 of the medicament delivery mechanism 4500 has a distal end portion 4402 and a proximal end portion 4404, and contains (i.e., is filled with or partially filled with) a naloxone composition 4420 (see, e.g., FIG. 16). The distal end portion 4402 of the medicament container 4400 contains a seal 4406. The seal 4406, which can be, for example, an 8-I crimp seal, is configured to burst when punctured by the proximal end 4456 of the needle 4452, as described below. The proximal end portion 4404 of the medicament container 4400 includes an elastomeric member 4410, and is configured to receive a piston portion 4534 of the movable member 4530. Although the medicament container 4400 is shown in FIG. 16 as including a liner 4407, in other embodiments, the medicament container 4400 need not include the liner 4407.

The medicament container 4400 can have any suitable size (e.g., length and/or diameter) and can contain any suitable volume of the naloxone composition 4420. Moreover, the medicament container 4400 and the movable member 4530 can be collectively configured such that the movable member 4530 travels a desired distance within the medicament container 4400 (i.e., the “stroke”) during an injection event. In this manner, the medicament container 4400, the volume of the naloxone composition 4420 within the medicament container 4400 and the movable member 4530 can be collectively configured to provide a desired fill volume and delivery volume. In some embodiments, for example, the size of the medicament container 4400 and the length of the movable member 4530 can be such that the fill volume of the naloxone composition 4420 is approximately 0.76 ml and the delivery volume of the naloxone composition 4420 is approximately 0.30 ml (providing a delivery volume to fill volume ratio of approximately 0.4). In other embodiments, for example, the size of the medicament container 4400 and the length of the movable member 4530 can be such that the fill volume of the naloxone composition 4420 is approximately 0.66 ml and the delivery volume of the naloxone composition 4420 is approximately 0.40 ml (providing a delivery volume to fill volume ratio of approximately 0.6).

Moreover, the length of the medicament container 4400 and the length of the movable member 4530 can be configured such that the medicament delivery mechanism 4500 can fit in the same housing 4110 regardless of the fill volume, the delivery volume and/or the ratio of the fill volume to the delivery volume. In this manner, the same housing and production tooling can be used to produce devices having various dosages of the naloxone composition. For example, in a first embodiment (e.g., having a fill volume to delivery volume ratio of 0.4), the medicament container has a first length and the movable member has a first length. In a second embodiment (e.g., having a fill volume to delivery volume ratio of 0.6), the medicament container has a second length shorter than the first length, and the movable member has a second length longer than the first length. In this manner, the stroke of the device of the second embodiment is longer than that of the device of the first embodiment, thereby allowing a greater dosage. The medicament container of the device of the second embodiment, however, is shorter than the medicament container of the device of the first embodiment, thereby allowing the components of both embodiments to be disposed within the same housing and/or a housing having the same length.

The naloxone composition 4420 contained within the medicament container 4400 can be any of the naloxone compositions described herein. In particular, the naloxone composition 4420 can include an effective amount of naloxone or salts thereof, a tonicity-adjusting agent, and a pH adjusting agent. The naloxone composition 4420 can be formulated such that the osmolality of the naloxone composition 4420 ranges from about 250-350 mOsm and the pH ranges from about 3-5.

In some embodiments, the naloxone composition 4420 can include any suitable concentration of 4,5-epoxy-3,14-dihydroxy-17-(2-propenyl)morphinan-6-one. In some embodiments, for example, the naloxone composition 4420 has a concentration of 4,5-epoxy-3,14-dihydroxy-17-(2-propenyl)morphinan-6-one between approximately 0.01 mg/mL, and approximately 10 mg/mL. In other embodiments, the naloxone composition 4420 has a concentration of 4,5-epoxy-3,14-dihydroxy-17-(2-propenyl)morphinan-6-one between approximately 0.05 and approximately 2 mg/mL.

The tonicity-adjusting agent can be any of the tonicity-adjusting agents described herein, and can be included within the naloxone composition 4420 in any suitable amount and/or concentration. For example, in some embodiments, the tonicity-adjusting agent includes at least one of dextrose, glycerin, mannitol, potassium chloride or sodium chloride. In other embodiments, the tonicity-adjusting agent includes sodium chloride in an amount such that a concentration of sodium chloride is between approximately 0.1 mg/mL and approximately 20 mg/mL.

The pH adjusting agent can be any of the pH adjusting agents described herein, and can be included within the naloxone composition 4420 in any suitable amount and/or concentration. For example, in some embodiments, the pH adjusting agent includes at least one of hydrochloric acid, citric acid, citrate salts, acetic acid, acetate salts, phosphoric acid or phosphate salts. In other embodiments, the pH adjusting agent includes a dilute hydrochloric acid.

The elastomeric member 4410 can be of any design or formulation suitable for contact with the naloxone composition 4420. For example, the elastomeric member 4410 can be formulated to minimize any reduction in the efficacy of the naloxone composition 4420 that may result from contact (either direct or indirect) between the elastomeric member 4410 and the naloxone composition 4420. For example, in some embodiments, the elastomeric member 4410 can be formulated to minimize any leaching or out-gassing of compositions that may have an undesired effect on the naloxone composition 4420. In other embodiments, the elastomeric member 4410 can be formulated to maintain its chemical stability, flexibility and/or sealing properties when in contact (either direct or indirect) with naloxone over a long period of time (e.g., for up to six months, one year, two years, five years or longer).

In some embodiments, the elastomeric member 4410 can be formulated to include a polymer and a curing agent. In such embodiments, the polymer can include at least one of bromobutyl or chlorobutyl. In such embodiments, the curing agent can include at least one of sulfur, zinc or magnesium.

In some embodiments, the elastomeric member 4410 can be constructed from multiple different materials. For example, in some embodiments, at least a portion of the elastomeric member 4410 can be coated. Such coatings can include, for example, polydimethylsiloxane. In some embodiments, at least a portion of the elastomeric member 4410 can be coated with polydimethylsiloxane in an amount of between approximately 0.02 mg/cm2 and approximately 0.80 mg/cm2.

As shown in FIG. 13, the system actuator 4300 includes the base 4302, a release member 4340 and a spring 4370. FIG. 14 shows certain of the internal components of the medical injector 4000 without the base 4302 and the spring 4370 so that the release member 4340 can be more clearly shown.

The release member 4340 has a proximal end portion 4342 and a distal end portion 4344, and is movably disposed within the distal end portion 4156 of the gas cavity 4154. The proximal end portion 4342 of the release member 4340 includes a sealing member 4345 and a puncturer 4341. The sealing member 4345 is configured to engage the sidewall of the housing 4110 defining the gas cavity 4154 such that the proximal end portion 4155 of the gas cavity 4154 is fluidically isolated from the distal end portion 4156 of the gas cavity 4154. In this manner, when gas is released from the gas container 4570, the gas contained in the proximal end portion 4155 of the gas cavity 4154 is unable to enter the distal end portion 4156 of the gas cavity 4154. The puncturer 4341 of the proximal end portion 4342 of the release member 4340 is configured to contact and puncture a frangible seal 4573 on the gas container 4570 when the release member 4340 moves proximally within the gas cavity 4154, as shown by the arrow BB in FIG. 14.

The distal end portion 4344 of the release member 4340 includes extensions 4343. The extensions 4343 include projections 4347 that include tapered surfaces 4349 and engagement surfaces 4348. Further, the extensions 4343 define an opening 4346 between the extensions 4343. The engagement surfaces 4348 of the projections 4347 are configured to extend through the safety lock aperture 4128 of the housing 4110 and contact a distal surface of the housing 4110, as shown in FIG. 15. In this manner, the engagement surfaces 4348 of the projections 4347 limit proximal movement of the release member 4340 when the engagement surfaces 4348 are in contact with the distal surface of the housing 4110.

The opening 4346 defined by the extensions 4343 is configured to receive the safety lock protrusion 4742 of the safety lock 4700 (see e.g., FIGS. 15 and 27). The safety lock protrusion 4742 is configured to prevent the extensions 4343 from moving closer to each other. Said another way, the safety lock protrusion 4742 is configured to ensure that the extensions 4343 remain apart and the engagement surfaces 4348 of the projections 4347 remain in contact with the distal end portion 4120 of the housing 4110. In some embodiments, for example, the release member 4340 and/or the extensions 4343 can be constructed from any suitable material configured to withstand deformation that may occur when exposed to a load over an extended period of time. In some embodiments, for example, the release member 4340 and/or the extensions 4343 can be constructed from brass.

The tapered surfaces 4349 of the projections 4347 are configured to contact protrusions 4313 on a proximal surface 4310 of the base 4302 (see e.g., FIG. 35) when the base 4302 is moved proximally relative to the housing 4110. Accordingly, when the base 4302 is moved proximally relative to the housing 4110, the extensions 4343 are moved together by the contact protrusions 4313. The inward movement of the extensions 4343 causes the release member 4340 to become disengaged from the distal end portion of the housing 4110, thereby allowing the release member 4340 to be moved proximally along its longitudinal axis as the spring 4370 expands.

The medicament delivery mechanism 4500 includes a gas container 4570, a carrier 4520, a movable member 4530, and a retraction spring 4590. As described above, the carrier 4520 and the movable member 4530 are disposed within the medicament cavity 4157 of the housing 4110. The gas container 4570 is disposed within the gas cavity 4154 of the housing 4110.

The gas container 4570 includes a distal end portion 4572 and a proximal end portion 4576, and is configured to contain a pressurized gas. The distal end portion 4572 of the gas container 4570 contains a frangible seal 4573 configured to break when the puncturer 4341 of the proximal end portion 4342 of the release member 4340 contacts the frangible seal 4573. The gas container retention member 4580 of the proximal cap 4112 of the housing 4110 is configured to receive and/or retain the proximal end portion 4576 of the gas container 4570. Said another way, the position of the gas container 4570 within the gas cavity 4154 is maintained by the gas container retention member 4580.

The movable member 4530 of the medicament delivery mechanism 4500 is movably disposed within the medicament cavity 4157. The movable member 4530 includes a piston portion 4534 having a plunger at the distal end portion of the piston portion 4534. The piston portion 4534 is configured to move within the medicament container 4400. In this manner, the piston portion 4534 of the movable member 4530 can apply a force to the elastomeric member 4410 to convey the naloxone composition 4420 contained in the medicament container 4400. The piston portion 4534 can be constructed of a resilient, durable, and/or sealing material, such as a rubber.

The carrier 4520 of the medicament delivery mechanism 4500 includes a distal end portion 4522 and a proximal end portion 4526. The medicament container 4400 is coupled to the carrier 4520 via a “snap-fit” connection (not shown) such that the medicament container 4400 can move relative to the carrier 4520 between a first configuration and a second configuration during an injection event. In the first configuration, the carrier 4520 is configured to move within the medicament cavity 4157 such that movement of the carrier 4520 within the medicament cavity 4157 causes contemporaneous movement of the medicament container 4400 within the medicament cavity 4157. The proximal end portion 4456 of the needle 4452 is spaced apart from the seal 4406 of the medicament container 4400 when the carrier 4520 and the medicament container 4400 are collectively in the first configuration (e.g., during needle insertion). When the carrier 4520 and the medicament container 4400 are moved to the second configuration, the medicament container 4400 releases from the “snap-fit” causing the medicament container 4400 to move distally with respect to the carrier 4520, causing the proximal end portion 4456 of the needle 4452 to pierce the seal 4406. In this manner, the needle 4452 can be selectively placed in fluid communication with the medicament container 4400 to define a medicament delivery path (not shown).

As shown in FIGS. 13, 14 and 41, the proximal end portion 4526 of the carrier 4520 includes a gas valve actuator 4527. The gas valve actuator 4527 is configured to engage a gas relief valve (not shown) of the movable member 4530 to allow the pressurized gas contained within the gas chamber (i.e., the volume within the medicament cavity 4157 between the proximal end of the housing 4110 and the proximal end of the movable member 5530) to escape when the injection event is complete. Thus, after the gas pressure within the medicament cavity 4157 decreases below a certain level, the force exerted by the retraction spring 4590 on the carrier 4520 can be sufficient to cause the carrier 4520 to move proximally within the housing 4110 (i.e., to retract). In addition, this arrangement results in there being substantially no residual force within the housing, which decreases stress on the components after the injection event.

FIGS. 18-28 show the electronic circuit system 4900. The electronic circuit system 4900 of the medical injector 4000 includes an electronic circuit system housing 4170, a printed circuit board 4922, a battery assembly 4962, an audio output device 4956, two light emitting diodes (LEDs) 4958A, 4958B and a battery clip 4910. As shown in FIG. 25, the electronic circuit system 4900 is configured to fit within the electronic circuit system cavity 4153 of the housing 4110. Accordingly, as described above, the electronic circuit system 4900 is physically and/or fluidically isolated from the medicament cavity 4157, the gas cavity 4154 and/or the medicament delivery device 4500. As described herein, the electronic circuit system 4900 is configured to output an electronic output associated with the use of the medical injector 4000.

The electronic circuit system housing 4170 of the electronic circuit system 4900 includes a distal end portion 4180 and a proximal end portion 4190. The proximal end portion 4190 includes connection protrusions 4171A and a battery clip protrusion 4173. The connection protrusions 4171A extend from the proximal end portion 4190 of the electronic circuit system housing 4170, and are configured to be disposed within the connection apertures 4152 of the housing 4110, as described above. In this manner, the electronic circuit system 4900 can be coupled to the housing 4110 within the electronic circuit system cavity 4153. In other embodiments, the electronic circuit system 4900 can be coupled to the housing 4110 by other suitable means such as an adhesive, a clip, a label and/or the like. As described in more detail herein, the battery clip protrusion 4173 is configured to hold the battery clip 4910 in place.

The proximal end portion 4190 of the electronic circuit system housing 4170 defines multiple sound apertures 4191. The audible output device 4956 is disposed against the proximal end portion 4190 of the electronic circuit system housing 4170 such that the front face of the audible output device 4956 is disposed adjacent the sound apertures 4191. In this manner, the sound apertures 4191 are configured to allow sound from an audio output device 4956 to pass from the audio output device 4956 to a region outside of the housing 4110.

As shown in FIGS. 21 and 22, the distal end portion 4180 of the electronic circuit system housing 4170 includes a connection protrusion 4171B, a stiffening protrusion 4174, and defines an LED aperture 4181, an aperture 4172, a safety lock actuator groove 4182, and a base actuator groove 4183. The LED aperture 4181 is configured to receive the LEDs 4958A, 4958B such that a user can view the LEDs 4958A, 4958B, which are described in more detail herein.

The connection protrusion 4171B extends from the distal end portion 4180 of the electronic circuit system housing 4170, and is configured to attach the electronic circuit system 4900 to the housing 4110, as described above. The stiffening protrusion 4174 is configured to have at least a portion received within and/or accessible via the aperture 4145 in the housing 4110 (see e.g., FIG. 7). The stiffening protrusion 4174 is configured to limit the bending (e.g., buckling) of the electronic circuit system housing 4170 when the electronic circuit system housing 4170 is coupled to the housing 4110. Moreover, a user can access the stiffening protrusion 4174 via the aperture 4172. In this manner, for example, the user can disengage the stiffening protrusion 4174 from the aperture 4145.

The safety lock actuator groove 4182 of the electronic circuit system housing 4170 is configured to be disposed adjacent the safety lock actuator groove 4123 of the distal end portion 4120 of the housing 4110. In this manner, the safety lock actuator groove 4182 of the electronic circuit system housing 4170 and the safety lock actuator groove 4123 of the distal end portion 4120 of the housing 4110 collectively receive the actuator 4744 of the safety lock 4700, which is described in more detail herein. Similarly, the base actuator groove 4183 of the electronic circuit system housing 4170 is configured to be disposed about the base actuator groove 4124 of the distal end portion 4120 of the housing 4110. The base actuator groove 4183 of the electronic circuit system housing 4170 and the base actuator groove 4124 of the distal end portion 4120 of the housing 4110 collectively receive the actuator 4311 of the base 4302, which is described in more detail herein.

The printed circuit board 4922 of the electronic circuit system 4900 includes a substrate 4924, a first actuation portion 4926 and a second actuation portion 4946. The substrate 4924 of the printed circuit board 4922 includes the electrical components necessary for the electronic circuit system 4900 to operate as desired. For example, the electrical components can be resistors, capacitors, inductors, switches, microcontrollers, microprocessors and/or the like. The printed circuit board may also be constructed of materials other than a flexible substrate such as a FR4 standard board (rigid circuit board).

As shown in FIGS. 26-28, the first actuation portion 4926 includes a first electrical conductor 4934 and defines an opening 4928 having a boundary 4929. The opening 4928 of the first actuation portion 4926 is configured to receive a protrusion 4746 of the actuator 4744 of the safety lock 4700. The boundary 4929 of the first opening 4928 has a discontinuous shape, such as, for example, a teardrop shape, that includes a stress concentration riser 4927. The discontinuity and/or the stress concentration riser 4927 of the boundary 4929 can be of any suitable shape to cause the substrate 4924 to deform in a predetermined direction when the protrusion 4746 of the actuator 4744 of the safety lock 4700 is moved relative to the opening 4928, as shown by the arrow CC in FIG. 27.

The opening 4928 is defined adjacent the first electrical conductor 4934 that electronically couples the components included in the electronic circuit system 4900. The first electrical conductor 4934 includes a first switch 4972, which can be, for example a frangible portion of the first electrical conductor 4934. In use, when the safety lock 4700 is moved from a first position (see e.g., FIG. 26) to a second position (see e.g., FIG. 27), the actuator 4744 moves in a direction substantially parallel to a plane defined by a surface of the first actuation portion 4926 of the substrate 4924. The movement of the actuator 4744 causes the protrusion 4746 to move within the first opening 4928, as indicated by the arrow CC in FIG. 27. The movement of the protrusion 4746 tears the first actuation portion 4926 of the substrate 4924, thereby separating the portion of the first electrical conductor 4934 including the first switch 4972. Said another way, when the safety lock 4700 is moved from its first position to its second position (see e.g., FIG. 38), the actuator 4744 moves irreversibly the first switch 4972 from a first state (e.g., a state of electrical continuity) to a second state (e.g., a state of electrical discontinuity). Said yet another way, when the safety lock 4700 is moved from its first position to its second position, the actuator 4744 disrupts the first electrical conductor 4934.

The second actuation portion 4946 includes a second electrical conductor 4935 and defines an opening 4945, having a boundary 4949 and a tear propagation limit aperture 4948. As shown in FIGS. 25-28, the opening 4945 of the second actuation portion 4946 is configured to receive a portion of an actuator 4311 of the base 4302. The boundary 4949 of the opening 4945 has a discontinuous shape that includes a stress concentration riser 4947. The discontinuity and/or the stress concentration riser 4947 of the boundary 4949 can be of any suitable shape to cause the substrate 4924 to deform in a predetermined direction when the actuator 4311 of the base 4302 is moved in a proximal direction relative to the opening 4945, as shown by the arrow DD in FIG. 23.

The second electrical conductor 4935 includes a second switch 4973 disposed between the opening 4945 and the tear propagation limit aperture 4948, which can be, for example, a frangible portion of the second electrical conductor 4935. In use, when the base 4302 is moved from its first position to its second position (see e.g., FIG. 39), the actuator 4311 moves in a proximal direction, substantially parallel to a plane defined by a surface of the second actuation portion 4946 of the substrate 4924. The proximal movement of the actuator 4311 tears the second actuation portion 4946 of the substrate 4924, thereby separating the portion of the second electrical conductor 4935 including the second switch 4973. Said another way, when the base 4302 is moved from its first position to its second position, the actuator 4311 moves irreversibly the second switch 4973 from a first state (e.g., a state of electrical continuity) to a second state (e.g., a state of electrical discontinuity). The tear propagation limit aperture 4948 is configured to limit the propagation of the tear in the substrate 4924 in the proximal direction. Said another way, the tear propagation limit aperture 4948 is configured to ensure that the tear in the substrate 4924 does not extend beyond the tear propagation limit aperture 4948. The tear propagation limit aperture 4948 can be any shape configured to stop the propagation of a tear and/or disruption of the substrate 4924. For example, the tear propagation limit aperture 4948 can be oval shaped. In other embodiments, the proximal boundary of the tear propagation limit aperture 4948 can be reinforced to ensure that the tear in the substrate 4924 does not extend beyond the tear propagation limit aperture 4948.

In some embodiments, the safety lock 4700 and base 4302 can be configured to interact with mechanical and/or optical switches to produce an electronic output in a reversible manner.

The battery assembly 4962 of the electronic circuit system 4900 includes two batteries stacked on top of one another. In other embodiments, the electronic circuit system can include any number of batteries and/or any suitable type of power source. In some embodiments, for example, the battery assembly can include Lithium batteries such as, for example, CR1616, CR2016s, type AAA or the like. The battery assembly 4962 has a first surface 4964 and a second surface 4966. The first surface 4964 of the battery assembly 4962 can contact an electrical contact (not shown) disposed on the substrate 4924. The second surface 4966 of the battery assembly 4962 is configured to contact a contact portion 4918 of a distal end portion 4916 of a battery clip 4910. When both the electrical contact of the substrate 4924 and the contact portion 4918 of the distal end portion 4916 of the battery clip 4910 contact the battery assembly 4962, the batteries of the battery assembly 4962 are placed in electrical communication with the electronic circuit system 4900. Said another way, when the electrical contact of the substrate 4924 and the contact portion 4918 of the distal end portion 4916 of the battery clip 4910 contact the battery assembly 4962, the battery assembly 4962 is configured to supply power to the electronic circuit system 4900.

The battery clip 4910 (shown in FIG. 23) includes a proximal end portion 4912 and a distal end portion 4916. The proximal end portion 4912 defines a retention aperture 4913. The retention aperture 4913 is configured to receive the battery clip protrusion 4173 of the electronic circuit system housing 4170. In this manner, the battery clip protrusion 4173 maintains the position of the battery clip 4910 with respect to the electronic circuit system housing 4170 and/or the battery assembly 4962.

The distal end portion 4916 of the battery clip 4910 includes a contact portion 4918 and an angled portion 4917. As described above, the contact portion 4918 is configured to contact the second surface 4966 of the battery assembly 4962 to place the battery assembly 4962 in electrical communication with the electronic circuit system 4900. The angled portion 4917 of the distal end portion 4916 of the battery clip 4910 is configured to allow a proximal end portion 4236 of a battery isolation protrusion 4235 (see e.g., FIG. 30) to be disposed between the second surface 4966 of the battery assembly 4962 and the contact portion 4918 of the distal end portion 4916 of the battery clip 4910. When the battery isolation protrusion 4235 is disposed between the second surface 4966 of the battery assembly 4962 and the contact portion 4918 of the distal end portion 4916 of the battery clip 4910, the electrical path between the battery assembly 4962 and the remainder of the electrical circuit system 4900 is severed, thereby removing power from the electronic circuit system 4900. The contact portion 4918 of the distal end portion 4916 of the battery clip 4910 is biased such that when the battery isolation protrusion 4235 is removed, the contact portion 4918 will move into contact the second surface 4966 of the battery assembly 4962, thereby restoring electrical communication between the battery assembly 4962 and the electronic circuit system 4900. In some embodiments, the battery isolation protrusion 4235 can be repeatedly removed from between the second surface 4966 of the battery assembly 4962 and the contact portion 4918 of the distal end portion 4916 of the battery clip 4910 and reinserted. Said another way, the battery isolation protrusion 4235 and the battery clip 4910 collectively form a reversible on/off switch.

The audio output device 4956 of the electronic circuit system 4900 is configured to output audible sound to a user in response to a use of the medical injector 4000. In some embodiments, the audible output device 4956 can be a speaker. In some embodiments, the audible sound can be, for example, associated with a recorded message and/or a recorded speech. In other embodiments, the audible instructions can be an audible beep, a series of tones and/or or the like.

In other embodiments, the medical injector 4000 can have a network interface device (not shown) configured to operatively connect the electronic circuit system 4900 to a remote device (not shown) and/or a communications network (not shown). In this manner, the electronic circuit system 4900 can send information to and/or receive information from the remote device. The remote device can be, for example, a remote communications network, a computer, a compliance monitoring device, a cell phone, a personal digital assistant (PDA) or the like. Such an arrangement can be used, for example, to download replacement processor-readable code from a central network to the electronic circuit system 4900. In some embodiments, for example, the electronic circuit system 4900 can download information associated with a medical injector 4000, such as an expiration date, a recall notice, updated use instructions or the like. Similarly, in some embodiments, the electronic circuit system 4900 can upload compliance information associated with the use of the medical injector 4000 via the network interface device.

FIGS. 24 and 25 show the cover 4200 of the medical injector 4000. The cover 4200 includes a proximal end portion 4210 and a distal end portion 4230, and defines a cavity 4242. The cavity 4242 of the cover 4200 is configured to receive at least a portion of the housing 4110. Thus, when the portion of the housing 4110 is disposed within the cover 4200, the cover 4200 blocks an optical pathway between the medicament container 4400 and a region outside of the housing 4110. Similarly stated, when the portion of the housing 4110 is disposed within the cover 4200, the cover 4200 is obstructs the first status indicator aperture 4150 and/or the second status indicator aperture 4151 of the housing 4110 to reduce the amount of light transmitted to the naloxone composition 4420 within the medicament container 4400. In this manner, the life of the naloxone composition 4420 can extended by the prevention and/or reduction of degradation to the naloxone that may be caused by ultra-violet radiation.

The proximal end portion 4210 of the cover 4200 defines apertures 4215 configured to receive the cover retention protrusions 4142 of the housing 4110 (shown in FIGS. 5 and 7). In this manner, the apertures 4215 and the cover retention protrusions 4142 of the housing 4110 removably retain the cover 4200 about at least a portion of the housing 4110. Said another way, the apertures 4215 and the cover retention protrusions 4142 of the housing 4110 are configured such that the cover 4200 can be removed from a portion of the housing 4110 and then replaced about the portion of the housing 4110.

As described above, the electronic circuit system 4900 can be actuated when the housing 4110 is at least partially removed from the cover 4200. More particularly, the distal end portion 4230 of the cover 4200 includes a battery isolation protrusion 4235. The battery isolation protrusion 4235 includes a proximal end portion 4236 and a tapered portion 4237. The proximal end portion 4236 of the battery isolation protrusion 4235 is configured to be removably disposed between the second surface 4966 of the battery assembly 4962 and the contact portion 4918 of the distal end portion 4916 of the battery clip 4910, as described above.

FIGS. 31-34 show the safety lock 4700 of the medical injector 4000. The safety lock 4700 of the medical injector 4000 includes a proximal surface 4740, a distal surface 4760 opposite the proximal surface 4740 and a needle sheath 4720. The safety lock 4700 defines a needle sheath aperture 4770 and a battery isolation protrusion aperture 4775. The battery isolation protrusion aperture 4775 is configured to receive the battery isolation protrusion 4235 of the cover 4200 such that the battery isolation protrusion 4235 can be disposed within the electronic circuit system cavity 4153 or the electronic circuit system 4900, as described above. Similarly stated, the battery isolation protrusion aperture 4775 of the safety lock 4700 is aligned with the battery isolation protrusion aperture 4121 of the housing 4110, such that the battery isolation protrusion 4235 can be disposed within the electronic circuit system cavity 4153 when the cover 4200 is disposed about a portion of the housing 4110.

The proximal surface 4740 of the safety lock 4700 includes a safety lock protrusion 4742, a stopper 4743, an actuator 4744 and two opposing pull tabs 4741. As described above, when the safety lock 4700 is in a first (locked) position, the safety lock protrusion 4742 is configured to be disposed in the opening 4346 defined by the extensions 4343 of the distal end portion 4344 of the release member 4340 (see also FIG. 15). Accordingly, the safety lock protrusion 4742 is configured to prevent the extensions 4343 from moving closer to each other, thereby preventing proximal movement of the release member 4340 of the medicament delivery mechanism 4500 and/or delivery of the naloxone composition 4420. The stopper 4743 of the safety lock 4700 is a protrusion extending from the proximal surface 4740 of the safety lock 4700. The stopper 4743 is configured to contact a portion of the housing 4110 to limit the proximal movement of the safety lock 4700 relative to the housing 4110. In other embodiments, the stopper 4743 can be any structure configured to limit the proximal movement of the safety lock 4700.

The actuator 4744 of the safety lock 4700 has an elongated portion 4745 and a protrusion 4746. The elongated portion 4745 extends in a proximal direction from the proximal surface 4740. In this manner, the elongated portion 4745 can extend through a safety lock actuator opening 4356 of the base 4302 (see e.g., FIG. 35) and within the safety lock actuator groove 4123 of the housing 4110 and the safety lock actuator groove 4182 of the electronic circuit system housing 4170. The protrusion 4746 extends in a direction substantially transverse to the elongated portion 4745 and/or substantially parallel to the proximal surface 4740 of the safety lock 4700. As described above, the opening 4928 of the first actuation portion 4926 is configured to receive the protrusion 4746 of the actuator 4744 of the safety lock 4700.

The pull tabs 4741 of the safety lock 4700 include a grip portion 4747 and indicia 4748. The grip portion 4747 of the pull tabs 4741 provides an area for the user to grip and/or remove the safety lock 4700 from the rest of the medicament delivery system 4700. The indicia 4748 provides instruction on how to remove the safety lock 4700. In some embodiments, for example, the indicia 4748 can indicate the direction the user should pull the safety lock 4700 to remove the safety lock 4700.

As shown in FIG. 33, the needle sheath 4720 of the safety lock 4700 includes a distal end portion 4724, a proximal end portion 4722 and a plurality of ribs 4728. The needle sheath 4720 can also define a lumen 4729. The lumen 4729 of the safety lock 4700 is configured to receive the needle 4452. In this manner, the needle sheath 4720 can protect the user from the needle 4452 and/or can keep the needle 4452 sterile before the user actuates the medical injector 4000. The proximal end portion 4722 of the needle sheath is configured to contact the distal end portion 4522 of the carrier 4520 of the medicament delivery mechanism 4500.

The distal end portion 4724 of the needle sheath 4720 has an angled ridge 4725. The angled ridge 4725 is configured to allow the proximal end portion 4722 of the needle sheath 4720 to irreversibly move through the needle sheath aperture 4770 of the safety lock 4700 in a distal direction. Said another way, the angled ridge 4725 can be configured in such a way as to allow the proximal end portion 4722 of the needle sheath 4720 to move through the needle sheath aperture 4770 in a distal direction, but not in a proximal direction. The needle sheath aperture 4770 has retaining tabs 4771 configured to engage the proximal end of the angled ridge 4725 when the needle sheath 4720 is moved in a proximal direction. In this manner, the retaining tabs 4771 prevent the proximal movement of the needle sheath with respect to the safety lock 4700. Further, the retaining tabs 4771 are configured to engage the proximal end of the angled ridge 4725 when the safety lock 4700 is moved in a distal direction. Said another way, as shown in FIG. 33, the needle sheath 4720 is removed from the needle 4452 when the safety lock 4700 is moved in a distal direction with respect to the housing 4110.

FIGS. 35 and 36 show the base 4302 of the medical injector 4000. The base 4302 includes a proximal surface 4310, a distal surface 4330 and base connection knobs 4358. The base 4302 defines a needle aperture 4350, a safety lock protrusion aperture 4352, a battery isolation protrusion aperture 4354, a safety lock actuator opening 4356, and pull tab openings 4360. The needle aperture 4350 is configured to receive the needle 4452 when the medical injector 4000 is actuated. The safety lock protrusion aperture 4352 of the base 4302 receives the safety lock protrusion 4742 of the safety lock 4700. The battery isolation protrusion aperture 4354 of the base 4302 receives the battery isolation protrusion 4235 of the cover 4200 and the stopper 4743 of the safety lock 4700. The safety lock actuator opening 4356 receives the safety lock actuator 4744 of the safety lock 4700. The pull tab openings 4360 are configured to receive the pull tabs 4741 of the safety lock 4700.

The proximal surface 4310 of the base 4302 includes an actuator 4311, guide members 4312, and protrusions 4313. The actuator 4311 is an elongate member configured to engage the substrate 4924 of the electronic circuit system 4900. As described above, the opening 4945 of the second actuation portion 4946 is configured to receive the actuator 4311 of the base 4302. The guide members 4312 of the base 4302 are configured to engage and/or slide within the base rail grooves 4127 of the housing 4110, as described above. The protrusions 4313 of the base 4302 are configured to engage the tapered surfaces 4349 of the extensions 4343 of the release member 4340. As described in further detail herein, when the safety lock 4700 is removed and the base 4302 is moved in a proximal direction with respect to the housing 4110, the protrusion 4313 of the base 4302 are configured to move the extensions 4343 of the release member 4340 closer to each other, actuating the medicament delivery mechanism 4500. As described above, the base connection knobs 4358 are configured to engage the base retention recesses 4125A, 4125B in a way that allows proximal movement of the base 4302 but limits distal movement of the base 4302.

As shown in FIG. 37, the medical injector 4000 is first enabled by moving the medicament delivery device 4000 from a first configuration to a second configuration by moving the cover 4200 from a first position to a second position. The cover 4200 is moved from the first position to the second position by moving it with respect to the housing 4110 in the direction shown by the arrow EE in FIG. 37. When the cover 4200 is moved with respect to the housing 4110 in the direction EE, the battery isolation protrusion 4235 is removed from the area between the battery clip 4910 and the second surface 4966 of the battery assembly 4962. In this manner, the battery assembly 4962 can be operatively coupled to the electronic circuit system 4900 when the cover 4200 is removed, thereby providing power to the electronic circuit system 4900. Similarly stated, this arrangement allows the electronic circuit system 4900 to be actuated when the cover 4200 is removed.

When power is provided, as described above, the electronic circuit system 4900 can output one or more predetermined electronic outputs. For example, in some embodiments, the electronic circuit system 4900 can output an electronic signal associated with recorded speech to the audible output device 4956. Such an electronic signal can be, for example, associated with a .WAV file that contains a recorded instruction instructing the user in the operation of the medical injector 4000. Such an instruction can state, for example, “remove the safety tab near the base of the auto-injector.” The electronic circuit system 4900 can simultaneously output an electronic signal to one and/or both of the LEDs 4958A, 4958B thereby causing one and/or both of the LEDs 4958A, 4958B to flash a particular color. In this manner, the electronic circuit system 4900 can provide both audible and visual instructions to assist the user in the initial operation of the medical injector 4000.

In other embodiments, the electronic circuit system 4900 can output an electronic output associated with a description and/or status of the medical injector 4000 and/or the naloxone composition 4420 contained therein. For example, in some embodiments, the electronic circuit system 4900 can output an audible message indicating the symptoms for which the naloxone composition should be administered, the expiration date of the naloxone composition, the dosage of the naloxone composition or the like.

As described above, the medical injector 4000 can be can be repeatedly moved between the first configuration and the second configuration when the cover 4200 is moved repeatedly between the first position and the second position respectively. Said another way, the cover 4200 can be removed and replaced about the housing 4110 any number of times. When the cover 4200 is moved from the second position to the first position, the battery isolation protrusion 4235 is inserted between the battery clip 4910 and the second surface 4966 of the battery assembly 4962, deactivating the electronic circuit system 4900. When the cover is moved from the first position to the second position a second time, the electronic circuit system 4900 is once again activated. In this manner, the cover 4200 can be removed and the electronic circuit system 4900 can output an electronic output without compromising the sterility of the needle 4452.

After the cover 4200 is removed from the housing 4110, the medical injector 4000 can be moved from the second configuration to a third configuration by moving the safety lock 4700 from a first position to a second position. The safety lock 4700 is moved from a first position to a second position by moving the safety lock 4700 with respect to the housing 4110 in the direction shown by the arrow FF in FIG. 38. When the safety lock 4700 is moved from the first position to the second position, the safety lock protrusion 4742 is removed from between the extensions 4343 of the release member 4340, thereby enabling the medicament delivery member 4500. Moreover, as shown in FIGS. 26 and 27, when the safety lock 4700 is moved from the housing 4110, the actuator 4744 of the safety lock 4700 moves in the direction CC as shown in FIG. 27, irreversibly moving the first switch 4972 from a first state (e.g., a state of electrical continuity) to a second state (e.g., a state of electrical discontinuity). When the actuator 4744 of the safety lock 4700 moves irreversibly the first switch 4972 of the electronic circuit system 4900 to the second state, the electronic circuit system 4900 can output one or more predetermined electronic outputs. For example, in some embodiments, a processor (not shown) can output an electronic signal associated with recorded speech to the audible output device 4956. Such an electronic signal can be, for example, associated with a recorded message notifying the user of the status of the medical injector 4000. Such a status message can state, for example, “If ready to use the naloxone medical injector, pull off the red safety guard.” The electronic circuit system 4900 can also simultaneously output an electronic signal to one and/or both of the LEDs 4958A, 4958B, thereby causing one and/or both of the LEDs 4958A, 4958B to stop flashing, change color or the like.

In some embodiments, the first actuation portion 4926 and the actuator 4744 can be configured such that the actuator 4744 must move a predetermined distance before the actuator 4744 engages the boundary 4929 of the opening 4928. For example, in some embodiments, the actuator 4744 must move approximately 0.200 inches before the actuator 4744 engages the boundary 4929 of the opening 4928. In this manner, the safety lock 4700 can be moved slightly without irreversibly moving the first switch 4972 of the electronic circuit system 4900 to the second state. Accordingly, this arrangement will permit the user to inadvertently and/or accidentally move the safety lock 4700 without actuating the electronic circuit system 4900.

In some embodiments, the electronic circuit system 4900 can be configured to output the status message for a predetermined time period, such as, for example, five seconds. After the predetermined time period has elapsed, the electronic circuit system 4900 can output an audible message further instructing the user in the operation of the medical injector 4000. Such an instruction can state, for example, “Place the base of the auto-injector against the patient's thigh. To complete the injection, press the base firmly against the patient's thigh.” In some embodiments, the electronic circuit system 4900 can simultaneously output an electronic signal to one and/or both of the LEDs 4958A, 4958B, thereby causing one and/or both of the LEDs 4958A, 4958B to flash a particular color. In this manner, the electronic circuit system 4900 can provide both audible and/or visual instructions to assist the user in the placement and actuation of the medical injector 4000. In some embodiments, the electronic circuit system 4900 can be configured to repeat the instructions after a predetermined time period has elapsed.

As described above, in other embodiments, the medical injector 4000 can have a network interface device (not shown) configured to operatively connect the electronic circuit system 4900 to a remote device (not shown) and/or a communications network (not shown). In this manner, the electronic circuit system 4900 can send a wireless signal notifying a remote device that the safety lock 4700 of the medical injector 4000 has been removed and that the medical injector 4000 has been armed. In other embodiments, the electronic circuit system 4900 can send a wireless signal (e.g., a wireless 911 call) notifying an emergency responder that the medical injector 4000 has been armed.

After the safety lock 4700 is moved from the first position to the second position, the medical injector 4000 can be moved from the third configuration to a fourth configuration by moving the base 4302 from a first position to a second position. Similarly stated, the medical injector 4000 can be actuated by the system actuator assembly 4300 by moving the base 4302 distally relative to the housing 4110. The base 4302 is moved from its first position to its second position by placing the medical injector 4000 against the body of the patient and moving the base 4302 with respect to the housing 4110 in the direction shown by the arrow GG in FIG. 39. Moving the base 4302 from the first position to the second position causes the protrusions 4313 on the proximal surface 4310 of the base 4302 to engage the tapered surfaces 4349 of the extensions 4343 of the release member 4340, thereby moving the extensions 4313 together. The inward movement of the extensions 4343 causes the release member 4340 to become disengaged from the distal end portion of the housing 4110, thereby allowing the release member 4340 to be moved proximally along its longitudinal axis as the spring 4370 expands.

When the base 4302 is moved from the first position to the second position, the system actuator 4300 actuates the medicament delivery mechanism 4500, thereby placing the medical injector 4500 in its fourth configuration (i.e., the needle insertion configuration), as shown in FIGS. 39 and 40. More particularly, when the medical injector is in its fourth configuration, the puncturer 4341 of the release member 4340 is in contact with and/or disposed through the frangible seal 4573 of the gas container 4570.

After the frangible seal 4573 has been punctured, an actuating portion of a compressed gas can escape from the gas container 4570 and flow via the gas passageway 4144 into the medicament cavity 4157. The gas applies gas pressure to the movable member 4530 causing the movable member 4530 and the carrier 4520 to move in a distal direction within the medicament cavity 4157, as shown by the arrow HH in FIG. 40. When the carrier 4520 moves distally within the medicament cavity 4157, the carrier 4520 and the medicament container 4400 are in a first configuration. Accordingly, as described above, the medicament container 4400 is connected to the carrier 4520 by a “snap fit” connection. In this manner, the medicament container 4400 and the needle 4452 contemporaneously move with movable member 4530 and/or the carrier 4520 in a distal direction. As described above, the proximal end portion 4456 of the needle 4452 is connected to the distal end portion 4522 of the carrier 4520 and is spaced from the seal 4406 of the medicament container 4400 when the carrier 4520 is in its first configuration. Said another way, the medicament container 4400 and the needle 4452 do not define a medicament delivery path when the carrier 4520 is in the first configuration. The movement of the needle 4452 in a distal direction causes the distal end portion of the needle 4452 to exit the housing 4110 and enter the body of a patient prior to administering the naloxone composition 4420.

After the carrier 4520 and/or the needle 4452 have moved within the medicament cavity 4157 a predetermined distance, the carrier 4520 and the medicament container 4400 are moved from the first configuration to a second configuration. In the second configuration of the carrier 4520, the medicament container 4400 is released from the “snap-fit” allowing the medicament container 4400 and the movable member 4530 to continue to move in a distal direction relative to the carrier 4520. Said another way, the medicament container 4400 is configured to slidably move within the carrier 4520 when the carrier is moved from the first configuration to the second configuration. As the medicament container 4400 continues to move within the carrier 4520, the proximal end portion 4456 of the needle 4452 contacts and punctures the seal 4406 of the medicament container 4400. This allows the medicament contained in the medicament container 4400 to flow into the lumen (not shown) defined by the needle 4452, thereby defining a medicament delivery path.

After the medicament container 4400 contacts the distal end of the carrier 4520, the medicament container 4400 stops moving within the carrier 4520 while the movable member 4530 continues to move in a distal direction, as shown by the arrow II in FIG. 41. This causes the piston portion 4534 of the movable member 4530 to move within the medicament container 4400 containing the naloxone composition 4420. As the piston portion 4534 of the movable member 4530 moves within the medicament container 4400, the piston portion 4534 contacts the elastomeric member 4410 and generates a pressure upon the naloxone composition 4420 contained within the medicament container 4400, thereby allowing at least a portion of the naloxone composition 4420 to flow out of the medicament container 4400 and into the lumen defined by the needle 4452. The medicament is delivered to a body of a user via the medicament delivery path defined by the medicament container 4400 and the needle 4452.

As shown in FIG. 42, after the movable member 4530 moves a predetermined distance within the medicament container 4400, the gas valve actuator 4527 of the carrier 4520 engages the gas relief valve (not shown in FIG. 42) of the movable member 4530 thereby allowing the pressurized gas contained within the gas chamber (i.e., the volume within the medicament cavity 4157 between the proximal end of the housing 4110 and the proximal end of the movable member 5530) to escape. Similarly stated, the gas valve actuator 4527 of the carrier 4520 engages the gas relief valve of the movable member 4530, the pressure within the housing 4110 is reduced, thereby ending the injection event. In this manner, the pre-injection distance between the proximal end portion of the movable member 4530 and the gas valve actuator 4527 of the carrier 4520 can be adjusted to control the amount of the naloxone composition 4420 to be injected. After the gas pressure within the medicament cavity 4157 decreases below a certain level, the force exerted by the retraction spring 4590 on the carrier 4520 can be sufficient to cause the carrier 4520 to move proximally within the housing 4110 (i.e., to retract), as shown by the arrow JJ in FIG. 42.

As described above, the actuator 4311 of the base 4302 actuates the electronic circuit 4900 to trigger a predetermined output or sequence of outputs when the base 4302 is moved from its first position to its second position (see, e.g., FIGS. 24-28). When the actuator 4311 is moved in a proximal direction relative to the opening 4945, as shown by the arrow DD in FIG. 28, the electronic circuit system 4900 is actuated to output one or more predetermined electronic outputs. For example, in some embodiments, the electronic circuit system 4900 can output an electronic signal associated with recorded speech to the audible output device 4956. Such an electronic signal can be, for example, associated with an audible countdown timer, instructing the user on the duration of the injection procedure. Said another way, if it takes, for example, ten seconds to complete an injection, an audible countdown timer can count from ten to zero ensuring that the user maintains the medical injector 4000 in place for the full ten seconds. In other embodiments, the electronic signal can be, for example, associated with a recorded message notifying the user that the injection is complete, instructing the user on post-injection disposal and safety procedures, instructing the user on post-injection medical treatment or the like. Such a status message can state, for example, “The injection is now complete. Please seek further medical attention from a doctor.” The electronic circuit system 4900 can also simultaneously output an electronic signal to one and/or both LEDs 4958A, 4958B, thereby causing one and/or both LEDs 4958A, 4958B to stop flashing, change color or the like, to provide a visual indication that the injection is complete. In other embodiments, the electronic circuit system 4900 can send a wireless signal notifying a remote device that the injection is complete. In this manner, a patient's compliance can be monitored.

In some embodiments, the second actuation portion 4946 and the actuator 4311 can be configured such that the base 4500 and/or the actuator 4311 must move a predetermined distance before the actuator 4311 engages the boundary 4949 of the opening 4945. For example, in some embodiments, the actuator 4311 must move approximately 0.200 inches before the actuator 4311 engages the boundary 4949 of the opening 4945. In this manner, the base 4700 can be moved slightly without irreversibly moving the second switch 4973 of the electronic circuit system 4900 to the second state. Accordingly, this arrangement will permit the user to inadvertently and/or accidentally move the base 4500 without actuating the electronic circuit system 4900.

Although the electronic circuit system 4900 is shown and described above as having two irreversible switches (e.g., switch 4972 and switch 4973), in other embodiments, an electronic circuit system can have any number of switches. Moreover, such switches can be either reversible or irreversible. For example, FIGS. 43-48 show portions of a medicament delivery device 5000 having an electronic circuit system 5900 having three irreversible switches.

The medicament delivery device 5000 is similar to the medical injector 4000 described above. As shown in FIGS. 43 and 44 (which show only portions of the medicament delivery device 5000), the medicament delivery device 5000 includes a housing 5110, a delivery mechanism (not shown), an electronic circuit system 5900, a cover (not shown), a safety lock (not shown, similar to safety lock 4700) and a base (not shown, similar to base 4302). The structure and operation of the delivery mechanism, the cover, the safety lock and the base are similar to the structure and operation of the delivery mechanism 4500, the cover 4200, the safety lock 4700 and the base 4302, respectively. Accordingly, only the electronic circuit system 5900 and the housing 5110 are described in detail below.

As shown in FIG. 43, the housing 5110 has a proximal end portion 5140 and a distal end portion 5120. The housing 5110 defines a gas cavity (not shown), a medicament cavity (not shown) and an electronic circuit system cavity 5153. The gas cavity and medicament cavity of the housing 5110 of the medicament delivery device 5000 are similar to the gas cavity 4154 and the medicament cavity 4157, shown and described above with reference to FIGS. 10 and 11.

The electronic circuit system cavity 5153 is configured to receive the electronic circuit system 5900. As described above, the electronic circuit system cavity 5153 is fluidically and/or physically isolated from the gas cavity and/or the medicament cavity by a sidewall 5148. The housing 5110 has protrusions 5149 configured to stabilize the electronic circuit system 5900 when the electronic circuit system 5900 is disposed within the electronic circuit system cavity 5153. The housing 5110 also defines connection apertures (not shown) configured to receive connection protrusions 5171 of the electronic circuit system 5900 (see e.g., FIG. 44). In this manner, the electronic circuit system 5900 can be coupled to the housing 5110 within the electronic circuit system cavity 5153 (see e.g., FIG. 47). In other embodiments, the electronic circuit system 5900 can be coupled within the electronic circuit system cavity 5153 by any other suitable means, such as an adhesive, a clip and/or the like.

The housing 5110 includes an actuation protrusion 5114 disposed within the electronic circuit system cavity 5153. As described in more detail herein, an angled end portion 5115 of the actuation protrusion 5114 of the housing 5110 is configured to engage a third actuation portion 5976 of a substrate 5924 of the electronic circuit system 5900 when the electronic circuit system 5900 is coupled to the housing 5110.

As shown in FIG. 47, the electronic circuit system 5900 is configured to fit within the electronic circuit system cavity 5153 of the housing 5110. Accordingly, as described above, the electronic circuit system 5900 is physically and/or fluidically isolated from the medicament cavity, the gas cavity and/or the medicament delivery path within the medicament delivery device 5000 (not shown). As described herein, the electronic circuit system 5900 is configured to output an electronic output associated with a use of the medicament delivery device 5000.

As shown in FIG. 44, the electronic circuit system 5900 is similar to the electronic circuit system 4900 described above. The electronic circuit system 5900 of the medicament delivery device 5000 includes an electronic circuit system housing 5170, a printed circuit board 5922, a battery assembly 5962, an audio output device 5956, two light emitting diodes (LEDs) 5958A, 5958B and a battery clip 5910. The electronic circuit system housing 5170, the battery assembly 5962, the audio output device 5956, the two light emitting diodes (LEDs) 5958A, 5958B and the battery clip 5910 are similar to the electronic circuit system housing 4170, the battery assembly 4962, the audio output device 4956, the two light emitting diodes (LEDs) 4958A, 4958B and the battery clip 4910 of the electronic circuit system 4900 described above. Thus, a detailed discussion of these components is omitted.

The electronic circuit system 5900 also includes a processor 5950 configured to process electronic inputs (e.g., from input switches) and produce electronic outputs. As described herein, such electronic outputs can include audio or visual outputs associated with a use of the medicament delivery device 5000. The processor 5950 can be a commercially-available processing device dedicated to performing one or more specific tasks. For example, in some embodiments, the processor 5950 can be a commercially-available microprocessor, such as the Sonix SNC 12060 or the SNC 26120 voice synthesizers. Alternatively, the processor 5950 can be an application-specific integrated circuit (ASIC) or a combination of ASICs, which are designed to perform one or more specific functions. In yet other embodiments, the processor 5950 can be an analog or digital circuit, or a combination of multiple circuits.

The processor 5950 can include a memory device (not shown) configured to receive and store information, such as a series of instructions, processor-readable code, a digitized signal, or the like. The memory device can include one or more types of memory. For example, the memory device can include a read only memory (ROM) component and a random access memory (RAM) component. The memory device can also include other types of memory suitable for storing data in a form retrievable by the processor 5950, for example, electronically-programmable read only memory (EPROM), erasable electronically-programmable read only memory (EEPROM), or flash memory.

FIG. 45 shows the printed circuit board 5922 of the electronic circuit system 5900. FIG. 46 is a schematic illustration of the electronic circuit system 5900. The printed circuit board 5922 of the electronic circuit system 5900 includes a substrate 5924, a first actuation portion 5926 (including a first switch 5972), a second actuation portion 5946 (including a second switch 5973), and a third actuation portion 5976 (including an electronic circuit system configuration switch 5974). The substrate 5924 of the printed circuit board 5922 includes the electrical components necessary for the electronic circuit system 5900 to operate as desired. For example, the electrical components can include resistors, capacitors, inductors, switches, microcontrollers, microprocessors and/or the like.

The first actuation portion 5926 and the second actuation portion 5946 are similar to the first actuation portion 4926 and the second actuation portion 4946 of the electronic circuit system 4900, described above (see e.g., FIG. 44), and are therefore not described or labeled in detail. The third actuation portion 5976 includes a third electrical conductor 5936 (see e.g., FIG. 45) and defines an actuation aperture 5975 having a boundary 5979, and a tear propagation limit aperture 5978. As shown in FIGS. 44 and 48, the actuation aperture 5975 of the third actuation portion 5976 is configured to receive the angled end portion 5115 of the actuation protrusion 5114 of the housing 5110 when the electronic circuit system 5900 is disposed within the electronic circuit system cavity 5153. The boundary 5979 of the actuation aperture 5975 has a discontinuous shape, such as, for example, a teardrop shape, that includes a stress concentration riser 5977. The discontinuity and/or the stress concentration riser 5977 of the boundary 5979 can be of any suitable shape to cause the substrate 5924 to deform in a predetermined direction when the angled end portion 5115 of the actuation protrusion 5114 of the housing 5110 is inserted into the actuation aperture 5975 (see e.g., FIG. 48), as described below.

The third electrical conductor 5936 includes the electronic circuit system configuration switch 5974 (see e.g., FIG. 45) disposed between the actuation aperture 5975 and the tear propagation limit aperture 5978, which can be, for example, a frangible portion of the third electrical conductor 5436. As shown in FIGS. 47 and 48, when the electronic circuit system 5900 is attached to the housing 5110, a portion of the angled portion 5115 of the actuation protrusion 5114 is disposed within the actuation aperture 5975 of the third actuation portion 5976, as shown by the arrow KK in FIG. 48. Continued movement of the angled portion 5115 of the actuation protrusion 5114 within the third actuation portion 5976 of the substrate 5924 causes the third actuation portion 5976 of the substrate 5924 to tear, thereby separating the portion of the third electrical conductor 5936 including the electronic circuit system configuration switch 5974. Said another way, when the electronic circuit system 5900 is attached to the housing 5110, the actuation protrusion 5114 moves irreversibly the electronic circuit system configuration switch 5974 from a first state (e.g., a state of electrical continuity) to a second state (e.g., a state of electrical discontinuity).

The tear propagation limit aperture 5978 is configured to limit the propagation of the tear in the substrate 5924. Said another way, the tear propagation limit aperture 5978 is configured to ensure that the tear in the substrate 5924 does not extend beyond the tear propagation limit aperture 5978. The tear propagation limit aperture 5978 can be any shape configured to limit the propagation of a tear and/or disruption of the substrate 5924. For example, the tear propagation limit aperture 5978 can be oval shaped. In other embodiments, the boundary of the tear propagation limit aperture 5978 can be reinforced to ensure that the tear in the substrate 5924 does not extend beyond the tear propagation limit aperture 5978. The angled end portion 5115 of the actuation protrusion 5114 ensures that the tear in the substrate 5924 propagates in the desired direction. Said another way, the angled end portion 5115 of the actuation protrusion 5114 ensures that the tear in the substrate 5924 occurs between the actuation aperture 5975 and the tear propagation limit aperture 5978.

When the actuation protrusion 5114 of the housing 5110 moves irreversibly the electronic circuit system configuration switch 5974 of the electronic circuit system 5900 from the first state to the second state, the electronic circuit system 5900 can be moved between a first configuration and a second configuration. For example, in some embodiments, irreversibly moving the electronic circuit system configuration switch 5974 of the electronic circuit system 5900 to the second state places the electronic circuit system 5900 in the second configuration such that when power is applied to the electronic circuit system 5900, the electronic circuit system 5900 recognizes that the medicament delivery device 5000 is a certain type of medicament delivery device and/or is in a certain configuration. In some embodiments, the housing can be devoid of the actuation protrusion 5114, thus the electronic circuit system configuration switch 5974 is maintained in its first state when the electronic circuit system 5900 is attached to the housing 5110. In this manner, the electronic circuit system configuration switch 5974 can enable the electronic circuit system 5900 to be used in different types and/or configurations of medicament delivery devices. The dual functionality of the electronic circuit system 5900 enables production of the same electronic circuit system 5900 for multiple devices, thereby permitting mass production and decreasing the cost of production of the electronic circuit system 5900.

For example, in some embodiments the electronic circuit system 5900 can be used in either an actual medicament delivery device or a simulated medicament delivery device. A simulated medicament delivery device can, for example, correspond to an actual medicament delivery device and can be used, for example, to train a user in the operation of the corresponding actual medicament delivery device.

The simulated medicament delivery device can simulate the actual medicament delivery device in any number of ways. For example, in some embodiments, the simulated medicament delivery device can have a shape corresponding to a shape of the actual medicament delivery device, a size corresponding to a size of the actual medicament delivery device and/or a weight corresponding to a weight of the actual medicament delivery device. Moreover, in some embodiments, the simulated medicament delivery device can include components that correspond to the components of the actual medicament delivery device. In this manner, the simulated medicament delivery device can simulate the look, feel and sounds of the actual medicament delivery device. For example, in some embodiments, the simulated medicament delivery device can include external components (e.g., a housing, a needle guard, a sterile cover, a safety lock or the like) that correspond to external components of the actual medicament delivery device. In some embodiments, the simulated medicament delivery device can include internal components (e.g., an actuation mechanism, a compressed gas source, a medicament container or the like) that correspond to internal components of the actual medicament delivery device.

In some embodiments, however, the simulated medicament delivery device can be devoid of a medicament and/or those components that cause the medicament to be delivered (e.g., a needle, a nozzle or the like). In this manner, the simulated medicament delivery device can be used to train a user in the use of the actual medicament delivery device without exposing the user to a needle and/or a medicament. Moreover, the simulated medicament delivery device can have features to identify it as a training device to prevent a user from mistakenly believing that the simulated medicament delivery device can be used to deliver a medicament. For example, in some embodiments, the simulated medicament delivery device can be of a different color than a corresponding actual medicament delivery device. Similarly, in some embodiments, the simulated medicament delivery device can include a label clearly identifying it as a training device.

The actuation of the medicament delivery device configuration switch 5974 can configure the electronic circuit system 5900 to output a different electronic output when the medicament delivery device 5000 is a simulated medical injector than when the medicament delivery device 5000 is an actual medical injector. Said yet another way, the electronic circuit system 5900 can be configured to output a first series of electronic outputs when the electronic circuit system configuration switch 5974 is in the first state and a second series of electronic outputs when the electronic circuit system configuration switch 5974 is in the second state. In this manner, the electronic circuit system configuration switch 5974 can enable the same electronic circuit system 5900 to be used in both simulated medicament delivery devices and actual medicament delivery devices. When used on an actual medicament delivery device, for example, the housing can be devoid of the actuation protrusion 5114. The dual functionality of the electronic circuit system 5900 can decrease the cost of production of the electronic circuit system 5900 of the medicament delivery device 5000.

In other embodiments, moving the electronic circuit system configuration switch 5974 to the second state can place the electronic circuit system 5900 in any number of different functional configurations. For example, moving the electronic circuit system configuration switch 5974 from the first state to the second state can indicate the type of medicament in the medicament container, the dosage of the medicament and/or the language of the audible electronic outputs output by the electronic circuit system 5900.

In still other embodiments, any number of electronic circuit system configuration switches can be used. For example, multiple switches can be used to configure the electronic circuit system 5900 to output usage instructions in any number of languages. For example, if an electronic circuit system contained three configuration switches (e.g., switches A, B and C), switch A can correspond to English instructions, switch B to Spanish instructions and switch C to German instructions. Further, moving both switch A and B to the second state might correspond to French instructions. In this manner, a single electronic circuit system 5900 can be configured to output instructions in multiple languages.

The needle 4452, as well as any other needles shown and described herein, can have any diameter and/or length to facilitate the injection of the naloxone composition 4420. For example, the needle can have a length suitable to penetrate clothing and deliver the naloxone via a subcutaneous injection and/or an intramuscular injection. In some embodiments, the needle 4452 (and any needle disclosed herein) can have a length of greater than 1 inch, greater than 1.5 inches, greater than 2 inches, greater than 2.5 inches or greater than 3 inches. In some embodiments, the needle 4452 (and any needle disclosed herein) can have a lumen diameter of approximately between 19 gauge and 31 gauge.

Although the medical injectors 4000 and 5000 are shown and described above as being auto-injectors configured to deliver the naloxone compositions described herein via injection through a needle (e.g., needle 4452), in other embodiments, a medicament delivery device can be configured to deliver the naloxone compositions described herein via any suitable delivery member, and in any suitable manner. For example, in some embodiments, a medicament delivery device can include a delivery member that delivers the naloxone composition into the body via inhalation and/or intranasal delivery.

For example, FIG. 49 is a schematic illustration of a medicament delivery device 6000 according to an embodiment that is configured to deliver a naloxone composition intranasally and/or via inhalation. The medicament delivery device 6000 includes a housing 6110, a medicament container 6400, a delivery member 6450 and an energy storage member 6570. The medicament container 6400 is at least partially disposed within the housing 6110, and contains (i.e., is filled or partially filled with) a naloxone composition 6420. The delivery member 6450 is coupled to the medicament container 6400, and, as described herein, is configured to delivery the naloxone composition from the medicament container 6400 intranasally and/or via inhalation. The energy storage member 6570 is disposed within the housing 6110, and is configured to produce a force F4 to deliver the naloxone composition 6420 (e.g., from the medicament container 6400 to a body).

The naloxone composition 6420 can be any of the naloxone compositions described herein. In particular, the naloxone composition 6420 can include an effective amount of naloxone or salts thereof, a tonicity-adjusting agent, and a pH adjusting agent. The naloxone composition 6420 can be formulated such that the osmolality of the naloxone composition 6420 ranges from about 250-350 mOsm and the pH ranges from about 3-5.

In some embodiments, the naloxone composition 6420 can include any suitable concentration of 4,5-epoxy-3,14-dihydroxy-17-(2-propenyl)morphinan-6-one. In some embodiments, for example, the naloxone composition 6420 has a concentration of 4,5-epoxy-3,14-dihydroxy-17-(2-propenyl)morphinan-6-one between approximately 0.01 mg/mL and approximately 60 mg/mL. In other embodiments, the naloxone composition 6420 has a concentration of 4,5-epoxy-3,14-dihydroxy-17-(2-propenyl)morphinan-6-one between approximately 0.05 nag/mL and approximately 2 mg/mL.

The tonicity-adjusting agent can be any of the tonicity-adjusting agents described herein, and can be included within the naloxone composition 6420 in any suitable amount and/or concentration. For example, in some embodiments, the tonicity-adjusting agent includes at least one of dextrose, glycerin, mannitol, potassium chloride or sodium chloride. In other embodiments, the tonicity-adjusting agent includes sodium chloride in an amount such that a concentration of sodium chloride is between approximately 0.1 mg/mL and approximately 20 mg/mL.

The pH adjusting agent can be any of the pH adjusting agents described herein, and can be included within the naloxone composition 6420 in any suitable amount and/or concentration. For example, in some embodiments, the pH adjusting agent includes at least one of hydrochloric acid, citric acid, citrate salts, acetic acid, acetate salts, phosphoric acid or phosphate salts. In other embodiments, the pH adjusting agent includes a dilute hydrochloric acid.

The medicament container 6400 can be any container suitable for storing the naloxone composition 6420. In some embodiments, the medicament container 6400 can be, for example, a pre-filled syringe, a pre-filled cartridge, a vial, an ampule or the like. In other embodiments, the medicament container 6400 can be a container having a flexible wall, such as, for example, a bladder. Although shown and described as being partially disposed within the housing 6110, in other embodiments, the medicament container 6400 can be disposed entirely within the housing 6110. Moreover, in some embodiments, the medicament container 6400 can be movably disposed within the housing 6110, such as, for example, in a manner similar to the medicament container 4400 shown and described above.

The delivery member 6450 is coupled to the medicament container 6400 and defines, at least in part, a flow path through which the naloxone composition 6420 can be delivered into a body. Although shown as being directly coupled to a distal end portion of the medicament container 6400, in other embodiments, the delivery member 6450 can be indirectly coupled to the medicament container 6400, (e.g., via the housing 6110).

Moreover, in some embodiments, the delivery member 6450 can be coupled to, but fluidically isolated from, the medicament container 6400 prior to actuation of the energy storage member 6570. In this manner, the medicament delivery device 6000 can be stored for extended periods of time while maintaining the sterility of the naloxone composition 6420 contained within the medicament container 6400, reducing (or eliminating) any leakage of the naloxone composition 6420 from the medicament container 6400 or the like. This arrangement also reduces and/or eliminates the assembly operations (e.g., the operation of coupling the delivery member 6450 to the medicament container 6400) before the medicament delivery device 6000 can be used to administer the naloxone composition 6200. In this manner, the medicament delivery device 6000 produces a quick and accurate mechanism for delivering the naloxone composition 6420. Similarly stated by reducing and/or eliminating the assembly operations prior to use, this arrangement reduces likelihood that performance of medicament delivery device 6000 and/or the delivery member 6450 will be compromised (e.g., by an improper coupling, a leak or the like).

In some embodiments, the delivery member 6450 can be coupled to the medicament container 6400 via a coupling member (not shown in FIG. 49) having similar functionality to the carrier 4520 shown and described above with respect to the medicament delivery device 4000. In such an embodiment, the medicament container 6400 and/or the delivery member 6450 can be configured to move relative to the coupling member when the energy storage member 6570 is actuated. Such movement can fluidically couple the delivery member 6450 and the medicament container 6400, thereby defining a flow path through which the naloxone composition 6420 can be delivered to the patient.

In some embodiments, the delivery member 6450 can enhance the delivery of the naloxone composition 6420 thereby improving the efficacy of the naloxone composition 6420. Similarly stated, in some embodiments, the delivery member 6450 can produce a flow of the naloxone composition 6420 having desired characteristics to enhance the absorption rate of the naloxone composition 6420, to minimize the delivery of the naloxone composition 6420 to regions of the body in which such delivery is less effective (e.g., the throat, etc.) or the like.

For example, in some embodiments, the delivery member 6450 can produce a controlled flow rate of the naloxone composition 6420. In such embodiments, the delivery member 6450 can include one or more flow orifices, a tortuous flow path or the like, to produce a desired pressure drop and/or to control the flow through the delivery member 6450. For example, in some embodiments, the delivery member 6450 can be configured to minimize excessive delivery of the naloxone composition 6420. For example, for intranasal applications, the delivery member 6450 can reduce the likelihood of excess deposition of the naloxone composition 6420 on the mucosal membrane, which can result in a portion of the naloxone composition 6420 being nonabsorbed (e.g., running out of the nose or into the throat).

In some embodiments, the delivery member 6450 can be configured to atomize the naloxone composition 6420 to produce a spray for intransal administration. For example, in some embodiments, the delivery member 6450 can produce an atomized spray of the naloxone composition having a desired spray geometry (e.g., spray angle and/or plume penetration) and/or droplet size distribution. In some embodiments, for example, the delivery member 6450 can include two chambers to allow substantially simultaneous deliver o the naloxone composition 6420 into both nostrils of a patient. Moreover, the delivery member 6450 can be cooperatively configured with the energy storage member 6570 to produce an atomized spray of the naloxone composition having a desired spray geometry and/or droplet size distribution. In this manner, the medicament delivery device 6000 can produce a consistent spray to enhance the efficacy of the naloxone composition 6420 under a wide variety of conditions.

In some embodiments, for example, the delivery member 6450 and the energy storage member 6570 can be cooperatively configured such that, when the energy storage member 6570 is actuated, the medicament delivery device 6000 produces an atomized spray of the naloxone composition 6420 having a substantial portion of the droplets therein having size distribution of between about 10 microns and about 20 microns. In this manner, the amount of the naloxone composition 6420 delivered to the lungs (e.g., the amount of smaller droplets that bypass the mucosal membrane) and/or the amount of the naloxone composition 6420 that runs into the throat (e.g., the amount of larger droplets) is minimized. In some embodiments, the delivery member 6450 and the energy storage member 6570 are cooperatively configured to produce a spray of the naloxone composition 6420 having a droplet size distribution wherein approximately 85 percent of the droplets have a size of between approximately 10 microns and 150 microns.

As described above, in some embodiments, the energy storage member 6570 is configured to “match” the delivery member 6450. Said another way, in some embodiments, the energy storage member 6570 is configured to produce the force F4 within a predetermined range to ensure the desired functionality of the delivery member 6450. Accordingly, the energy storage member 6570 can be any suitable device or mechanism that, when actuated, produces the desired force F4 to deliver the naloxone composition 6420 as described herein. By employing the energy storage member 6570 to produce the force F4, rather than relying on a user to manually produce the delivery force, the naloxone composition 6420 can be delivered into the body at the desired pressure and/or flow rate, and with the desired characteristics, as described above. Moreover, this arrangement reduces the likelihood of partial delivery (e.g., that may result if the user is interrupted or otherwise rendered unable to complete the delivery).

In some embodiments, the energy storage member 6570 can be a mechanical energy storage member, such as a spring, a device containing compressed gas, a device containing a vapor pressure-based propellant or the like. In other embodiments, the energy storage member 6570 can be an electrical energy storage member, such as a battery, a capacitor, a magnetic energy storage member or the like. In yet other embodiments, the energy storage member 6570 can be a chemical energy storage member, such as a container containing two substances that, when mixed, react to produce energy.

Although the medicament delivery device 6000 is shown and described above as including an energy storage member 6570, in other embodiments, a kit can include a medicament container containing a naloxone composition that is delivered by a manually-produced force. For example, FIG. 50 is a schematic illustration of a kit 7000 according to an embodiment. The kit 7000 includes a case 7202, a medicament container 7400 that contains (i.e., is filled or partially filled with) a naloxone composition 7420, and a delivery member 7450. The naloxone composition 7420 can be any of the naloxone compositions shown and described herein. The medicament container 7400 is movably disposed within the case 7202. More particularly, the medicament container 7400 can be removed from the case 7202 to deliver the naloxone composition 7420 contained therein.

Although the medicament container 7400 is shown as being substantially enclosed by and/or disposed within the case 7202, in other embodiments, the medicament container 7400 can be only partially enclosed by and/or disposed within the case 7202. In some embodiments, the case 7202 blocks an optical pathway between the medicament container 7400 and a region outside of the case 7202. Similarly stated, when the medicament container 7400 is disposed within the case 7202, the case 7202 is obstructs the medicament container 7400 to reduce the amount of light transmitted to the naloxone composition 7420 within the medicament container 7400.

The delivery member 7450, which can be a needle, an atomizer (e.g., for intranasal delivery, as described above), a mouthpiece or the like, is coupled to the medicament container 7400 and defines, at least in part, a flow path through which the naloxone composition 7420 can be delivered into a body. Although shown as being directly coupled to a distal end portion of the medicament container 7400, in other embodiments, the delivery member 7450 can be indirectly coupled to the medicament container 7400, (e.g., via the housing 7110).

Moreover, in some embodiments, the delivery member 7450 can be coupled to, but fluidically isolated from, the medicament container 7400 prior to actuation of the medicament container 7400 (e.g., by manually depressing a plunger, squeezing a trigger, or the like). In this manner, the medicament delivery device 7000 can be stored for extended periods of time while maintaining the sterility of the naloxone composition 7420 contained within the medicament container 7400, reducing (or eliminating) any leakage of the naloxone composition 7420 from the medicament container 7400 or the like. This arrangement also reduces and/or eliminates the assembly operations (e.g., the operation of coupling the delivery member 7450 to the medicament container 7400) before the medicament delivery device 7000 can be used to administer the naloxone composition 7200. In this manner, the medicament delivery device 7000 produces a quick and accurate mechanism for delivering the naloxone composition 7420. Similarly stated by reducing and/or eliminating the assembly operations prior to use, this arrangement reduces likelihood that performance of medicament delivery device 7000 and/or the delivery member 7450 will be compromised (e.g., by an improper coupling, a leak or the like).

In some embodiments, the delivery member 7450 can be coupled to the medicament container 7400 via a coupling member (not shown in FIG. 50) having similar functionality to the carrier 4520 shown and described above with respect to the medicament delivery device 4000. In such an embodiment, the medicament container 7400 and/or the delivery member 7450 can be configured to move relative to the coupling member when the medicament container 7400 is actuated. For example, in use, upon depressing a plunger to actuate the medicament container 7400, the coupling member can move relative to the medicament container 7400 before a substantial portion of the energy produced by movement of the plunger is exerted on the naloxone composition 7420. Such movement can fluidically couple the delivery member 7450 and the medicament container 7400, thereby defining a flow path through which the naloxone composition 7420 can be delivered to the patient.

In some embodiments, at least one of the medicament container 7400 and the case 7202 can include an electronic circuit system (not shown in FIG. 50) similar to the electronic circuit systems shown and described herein. In such embodiments, the electronic circuit system can be actuated when the medicament container is removed from the case 7202. Any suitable mechanism can be used to actuate the electronic circuit system when the medicament container 7400 is removed from the case 7202. Such mechanisms include those mechanisms disclosed in U.S. Patent Publication No. 2007/0129708, entitled “Devices, Systems and Methods for Medicament Delivery,” filed on Feb. 5, 2007, which is incorporated herein by reference in its entirety.

While various embodiments of the invention have been described above, it should be understood that they have been presented by way of example only, and not limitation. Where methods described above indicate certain events occurring in certain order, the ordering of certain events may be modified. Additionally, certain of the events may be performed concurrently in a parallel process when possible, as well as performed sequentially as described above.

Although the medical injector 4000 includes the electronic circuit system cavity 4153, the gas cavity 4154 and/or the medicament cavity 4157 that are shown and described as being fluidically and/or physically isolated from each other, in other embodiments, any of the electronic circuit system cavity 4153, the gas cavity 4154 and/or the medicament cavity 4157 can be fluidically coupled to and/or share a common boundary with each other. In some embodiments, for example, a housing can define a single cavity within which a medicament container, an energy storage member and an electronic circuit system are disposed.

Although the medical injector 4000 discloses a gas-powered delivery device, in other embodiments, any of the medicament delivery devices disclosed herein can include any suitable energy storage member. Such energy storage members can be, for example, a mechanical energy storage member, such as a spring, a device containing compressed gas, a device containing a vapor pressure-based propellant or the like. In other embodiments, the energy storage member can be an electrical energy storage member, such as a battery, a capacitor, a magnetic energy storage member or the like. In yet other embodiments, the energy storage member can be a chemical energy storage member, such as a container containing two substances that, when mixed, react to produce energy.

The medicament containers and/or medicament delivery devices disclosed herein can contain any suitable amount of any of the naloxone compositions disclosed herein. For example, in some embodiments, a medicament delivery device as shown herein can be a single-dose device containing an amount of the naloxone composition to be delivered of approximately 0.4 mg, 0.8 mg, 1 mg, 1.6 mg or 2 mg. As described above, the fill volume can be such that the ratio of the delivery volume to the fill volume is any suitable value (e.g., 0.4, 0.6 or the like). In some embodiments, an electronic circuit system can include “configuration switch” (similar to the configuration switch 5974 shown and described above) that, when actuated during the assembly of the delivery device, can select an electronic output corresponding to the dose contained within the medicament container.

Although the electronic circuit system 4900 is shown and described above as having two irreversible switches (e.g., switch 4972 and switch 4973), in other embodiments, an electronic circuit system can have any number of switches. Such switches can be either reversible or irreversible.

Although the electronic circuit system 4900 is shown and described above as producing an electronic output in response to the actuation of two switches (e.g., switch 4972 and switch 4973), in other embodiments, an electronic circuit system can produce an electronic output in response to any suitable input, command or prompt. Suitable input for prompting an output can include, for example, an audible input by the user (e.g., the user's response to a voice prompt produced by the electronic circuit system), an input from a “start button” depressed by the user, an input from a sensor (e.g., a proximity sensor, a temperature sensor or the like), movement of (e.g., shaking) of the medicament delivery device, or the like. In some embodiments, an electronic circuit system can include a microphone and/or a voice recognition module to detect a user's vocal input.

Although medical devices having two LEDs and an audio output device have been shown, in other embodiments the medical device might have any number of LEDs and/or audio output devices. Additionally, other types of output devices, such as haptic output devices, can be used. In some embodiments, outputs from an electronic circuit system can include, for example, an audible or visual output related to the naloxone composition (e.g., an indication of the expiration date, the symptoms requirement treatment with naloxone or the like), the use of the medicament delivery device, and/or post-administration procedures (e.g., a prompt to call 911, instructions for the disposal of the device or the like).

Although various embodiments have been described as having particular features and/or combinations of components, other embodiments are possible having a combination of any features and/or components from any of embodiments where appropriate. For example, any of the devices shown and described herein can include an electronic circuit system as described herein. For example, although the medicament delivery device 6000 shown in FIG. 49 is not shown as including an electronic circuit system, in other embodiments, a medicament delivery device similar to the device 6000 can include an electronic circuit system similar to the electronic circuit system 4900 shown and described above.

Any of the medicament containers described herein can include any of the elastomeric members described herein. For example, the medicament container 7400 can include an elastomeric member that is formulated to be compatible with the naloxone composition contained therein, similar to the elastomeric member 2410 shown and described above.

Any of the medicament containers described herein can contain any of the naloxone compositions and/or formulations described herein.

Claims

1. An apparatus, comprising:

a housing;
a medicament container disposed within the housing, the medicament container containing a naloxone composition;
an elastomeric member movably disposed within the medicament container, the elastomeric member formulated to be compatible with the naloxone composition, at least a portion of the elastomeric member including a silicone-based material; and
an energy storage member disposed within the housing, the energy storage member configured to produce a force that is exerted on the elastomeric member to deliver the naloxone composition,
wherein the naloxone composition includes naloxone or salts thereof, a tonicity-adjusting agent, and a pH adjusting agent, whereby at least one of (A) the osmolality of the naloxone composition ranges from about 250-350 mOsm or (B) the pH ranges from about 3-5.

2. The apparatus of claim 1, wherein the pH adjusting agent is present in the naloxone composition in an amount sufficient to cause the pH of the naloxone composition to be within the range from 3-5.

3. The apparatus of claim 1, wherein the naloxone composition includes 4,5-epoxy-3,14-dihydroxy-17-(2-propenyl) morphinan-6-one.

4. The apparatus of claim 1, wherein the tonicity-adjusting agent includes at least one of dextrose, glycerin, mannitol, potassium chloride or sodium chloride.

5. The apparatus of claim 1, wherein the tonicity-adjusting agent includes sodium chloride in an amount such that a concentration of sodium chloride is between approximately 0.1 mg/mL and approximately 20 mg/mL.

6. The apparatus of claim 1, wherein the elastomeric member is formulated to include a polymer and a curing agent, the polymer including at least one of bromobutyl or chlorobutyl, the curing agent including at least one of sulfur or a metal oxide.

7. The apparatus of claim 1, wherein at least a portion of the elastomeric member is coated with polydimethylsiloxane.

8. The apparatus of claim 1, wherein at least the portion of the elastomeric member is coated with polydimethylsiloxane in an amount of between approximately 0.02 mg/cm2 and approximately 0.80 mg/cm2.

9. The apparatus of claim 1, further comprising:

an electronic circuit system coupled to the housing, the electronic circuit system configured to produce an output when the electronic circuit system is actuated.

10. The apparatus of claim 1, further comprising:

a delivery member coupled to the medicament container, the naloxone composition configured to be delivered from the medicament container via the delivery member.

11. The apparatus of claim 10, wherein the delivery member is a needle defining a lumen therein having a size of between 19 gauge and 31 gauge.

12. The apparatus of claim 10, wherein the delivery member is an atomizer configured to atomize the naloxone composition.

13. An apparatus, comprising:

a housing;
a medicament container disposed within the housing, the medicament container containing a naloxone composition;
a plunger movably disposed within the medicament container, at least a portion of the plunger including polydimethylsiloxane such that the portion of the plunger is compatible with the naloxone composition; and
an energy storage member disposed within the housing, the energy storage member configured to produce a force that is exerted on the plunger to deliver the naloxone composition,
wherein the naloxone composition includes naloxone or salts thereof, a tonicity-adjusting agent, and a pH adjusting agent, whereby at least one of (A) the osmolality of the naloxone composition ranges from about 250-350 mOsm or (B) the pH ranges from about 3-5.

14. The apparatus of claim 13, wherein the plunger is an elastomeric member.

15. The apparatus of claim 13, wherein the plunger is an elastomeric member formulated to include a polymer and a curing agent, the polymer including at least one of bromobutyl or chlorobutyl, the curing agent including at least one of sulfur or a metal oxide.

16. The apparatus of claim 13, wherein at least the portion of the plunger is coated with polydimethylsiloxane in an amount of between approximately 0.02 mg/cm2 and approximately 0.80 mg/cm2.

17. The apparatus of claim 13, wherein the tonicity-adjusting agent includes at least one of dextrose, glycerin, mannitol, potassium chloride or sodium chloride.

18. An apparatus, comprising:

a housing;
a medicament container assembly disposed within the housing, the medicament container assembly including: a naloxone composition including a tonicity-adjusting agent and a pH adjusting agent, the pH of the naloxone composition ranges from about 3-5; a silicone-based material such that the medicament container assembly is compatible with the naloxone composition; and a plunger; and
an actuator moveably coupled to the housing, the actuator configured to cause a force to be exerted on the plunger to expel the naloxone composition from the medicament container assembly when the actuator is moved relative to the housing.

19. The apparatus of claim 18, wherein the pH of the naloxone composition ranges from about 3.2-3.6.

20. The apparatus of claim 18, wherein the medicament container assembly includes a seal, the apparatus further comprising:

a delivery member configured to be coupled to the medicament container assembly, the naloxone composition configured to be expelled from the medicament container assembly via the delivery member, the seal configured to fluidically isolate the delivery member from the medicament container assembly before the actuator is moved relative to the housing, the delivery member configured to be placed in fluid communication with the medicament container assembly when the plunger moves relative to the medicament container.

21. The apparatus of claim 20 wherein the delivery member is configured to atomize the naloxone composition.

22. An apparatus, comprising:

a medicament container containing a naloxone composition, the naloxone composition including naloxone or salts thereof, a tonicity-adjusting agent, and a pH adjusting agent, whereby the pH of the naloxone composition ranges from about 3-5;
an elastomeric member configured to be disposed within the medicament container and configured to be in contact with the naloxone composition, the elastomeric member including a silicone-based material;
a carrier including a needle, the carrier configured to receive at least a distal end portion of the medicament container such that an end portion of the needle is fluidically isolated from the medicament container when the medicament container is in a first position within the carrier, the end portion of the needle being in fluid communication with the medicament container when the medicament container is in a second position within carrier; and
a movable member configured to exert a force on the elastomeric member to move the medicament container from the first position to the second position, and to deliver the naloxone composition via the needle.

23. The apparatus of claim 22, further comprising:

an energy storage member disposed within the housing, the energy storage member configured to produce the force.

24. The apparatus of claim 22, wherein the elastomeric member is a flexible wall configured to exert a force on the naloxone composition, the naloxone composition being expelled from the medicament container when the force is exerted.

25. The apparatus of claim 22, wherein at least a portion of the elastomeric member includes polydimethylsiloxane.

26. The apparatus of claim 22, wherein the carrier includes an atomizer configured to atomize the naloxone composition.

Referenced Cited
U.S. Patent Documents
2960087 November 1960 Uytenbogaart
3055362 September 1962 Uytenbogaart
3115133 December 1963 Morando
3426448 February 1969 Sarnoff
3688765 September 1972 Gasaway
3768472 October 1973 Hodosh et al.
3795061 March 1974 Sarnoff et al.
3945379 March 23, 1976 Pritz et al.
4108177 August 22, 1978 Pistor
4186741 February 5, 1980 Cesaro
4226235 October 7, 1980 Sarnoff et al.
4284077 August 18, 1981 Wagner
4360019 November 23, 1982 Portner et al.
4424057 January 3, 1984 House
4441629 April 10, 1984 Mackal
4484910 November 27, 1984 Sarnoff et al.
4573976 March 4, 1986 Sampson et al.
4596556 June 24, 1986 Morrow et al.
4610666 September 9, 1986 Pizzino
4613328 September 23, 1986 Boyd
4617557 October 14, 1986 Gordon
4624660 November 25, 1986 Mijers et al.
4640686 February 3, 1987 Dalling et al.
4643721 February 17, 1987 Brunet
4666430 May 19, 1987 Brown et al.
4673657 June 16, 1987 Christian
4689042 August 25, 1987 Sarnoff et al.
4693708 September 15, 1987 Wanderer et al.
4755172 July 5, 1988 Baldwin
4781697 November 1, 1988 Slaughter
4782841 November 8, 1988 Lopez
4784652 November 15, 1988 Wikström
4795433 January 3, 1989 Sarnoff
4853521 August 1, 1989 Claeys et al.
4874382 October 17, 1989 Lindemann et al.
4894054 January 16, 1990 Miskinyar
4906235 March 6, 1990 Roberts
4906563 March 6, 1990 Singh et al.
4915695 April 10, 1990 Koobs
4941880 July 17, 1990 Burns
4959056 September 25, 1990 Dombrowski et al.
4968302 November 6, 1990 Schluter et al.
4983164 January 8, 1991 Hook et al.
5000736 March 19, 1991 Kaufhold, Jr. et al.
5024656 June 18, 1991 Gasaway et al.
5037306 August 6, 1991 van Schoonhoven
5038023 August 6, 1991 Saliga
5041088 August 20, 1991 Ritson et al.
5042977 August 27, 1991 Bechtold et al.
5062603 November 5, 1991 Smith et al.
5064413 November 12, 1991 McKinnon et al.
5071353 December 10, 1991 van der Wal
5085642 February 4, 1992 Sarnoff et al.
5092843 March 3, 1992 Monroe et al.
5096715 March 17, 1992 Sinclair
5104380 April 14, 1992 Holman et al.
5125898 June 30, 1992 Kaufhold, Jr. et al.
5167641 December 1, 1992 Schmitz
5199949 April 6, 1993 Haber et al.
5224936 July 6, 1993 Gallagher
5240146 August 31, 1993 Smedley et al.
5271527 December 21, 1993 Haber et al.
5281198 January 25, 1994 Haber et al.
5286258 February 15, 1994 Haber et al.
5298023 March 29, 1994 Haber et al.
5312326 May 17, 1994 Myers et al.
5314412 May 24, 1994 Rex
5314502 May 24, 1994 McNichols et al.
5343519 August 30, 1994 Feldman
5344407 September 6, 1994 Ryan
5354284 October 11, 1994 Haber et al.
5356376 October 18, 1994 Milijasevic et al.
5363842 November 15, 1994 Mishelevich et al.
5380281 January 10, 1995 Tomellini et al.
5383851 January 24, 1995 McKinnon, Jr. et al.
5383864 January 24, 1995 van den Heuvel
5394866 March 7, 1995 Ritson et al.
5399163 March 21, 1995 Peterson et al.
5405362 April 11, 1995 Kramer et al.
5417660 May 23, 1995 Martin
5451210 September 19, 1995 Kramer et al.
5466217 November 14, 1995 Myers et al.
5514135 May 7, 1996 Earle
5558679 September 24, 1996 Tuttle
5567160 October 22, 1996 Massino
5568555 October 22, 1996 Shamir
5569192 October 29, 1996 van der Wal
5584815 December 17, 1996 Pawelka et al.
5587381 December 24, 1996 Sinclair
5610992 March 11, 1997 Hickman
5615771 April 1, 1997 Hollister
5616132 April 1, 1997 Newman
5645534 July 8, 1997 Chanoch
5662612 September 2, 1997 Niehoff
5681291 October 28, 1997 Galli
5681292 October 28, 1997 Tober et al.
5692492 December 2, 1997 Bruna et al.
5695476 December 9, 1997 Harris
5697916 December 16, 1997 Schraga
5716338 February 10, 1998 Hjertman et al.
5728074 March 17, 1998 Castellano et al.
5772635 June 30, 1998 Dastur et al.
5779677 July 14, 1998 Frezza
5792190 August 11, 1998 Olson et al.
5800397 September 1, 1998 Wilson et al.
5805423 September 8, 1998 Wever et al.
5809997 September 22, 1998 Wolf
5813397 September 29, 1998 Goodman et al.
5813570 September 29, 1998 Fuchs et al.
5814020 September 29, 1998 Gross
5823346 October 20, 1998 Weiner
5823363 October 20, 1998 Cassel
5832488 November 3, 1998 Eberhardt
5837546 November 17, 1998 Allen et al.
RE35986 December 8, 1998 Ritson et al.
5846089 December 8, 1998 Weiss et al.
5848988 December 15, 1998 Davis
5852590 December 22, 1998 de la Huerga
5853292 December 29, 1998 Eggert et al.
5858001 January 12, 1999 Tsals et al.
5865795 February 2, 1999 Schiff et al.
5866154 February 2, 1999 Bahal et al.
5868713 February 9, 1999 Klippenstein
5868721 February 9, 1999 Marinacci et al.
D407487 March 30, 1999 Greubel et al.
5925021 July 20, 1999 Castellano et al.
5928195 July 27, 1999 Malamud et al.
5941857 August 24, 1999 Nguyen et al.
5964739 October 12, 1999 Champ
5970457 October 19, 1999 Brant et al.
5971953 October 26, 1999 Bachynsky
5991655 November 23, 1999 Gross et al.
6015438 January 18, 2000 Shaw
6030363 February 29, 2000 Kriesel
6039713 March 21, 2000 Botich et al.
6045534 April 4, 2000 Jacobsen et al.
6050977 April 18, 2000 Adams
6056728 May 2, 2000 von Schuckmann
6062901 May 16, 2000 Liu et al.
6063053 May 16, 2000 Castellano et al.
6074213 June 13, 2000 Hon
6077106 June 20, 2000 Mish
6083199 July 4, 2000 Thorley et al.
6084526 July 4, 2000 Blotky et al.
6086562 July 11, 2000 Jacobsen et al.
6096002 August 1, 2000 Landau
6099504 August 8, 2000 Gross et al.
6102896 August 15, 2000 Roser
6119684 September 19, 2000 Nöhl et al.
6149626 November 21, 2000 Rachynsky et al.
6158613 December 12, 2000 Novosel et al.
6161281 December 19, 2000 Dando et al.
6165155 December 26, 2000 Jacobsen et al.
6175752 January 16, 2001 Say
6179812 January 30, 2001 Botich et al.
6192891 February 27, 2001 Gravel et al.
6193695 February 27, 2001 Rippstein, Jr.
6202642 March 20, 2001 McKinnon et al.
6210359 April 3, 2001 Patel et al.
6210369 April 3, 2001 Wilmot et al.
6219587 April 17, 2001 Ahlin et al.
6221045 April 24, 2001 Duchon et al.
6221055 April 24, 2001 Shaw et al.
6245046 June 12, 2001 Sibbitt
6258063 July 10, 2001 Haar et al.
6259654 July 10, 2001 de la Huerga
6264629 July 24, 2001 Landau
6270455 August 7, 2001 Brown
6270479 August 7, 2001 Bergens et al.
6284765 September 4, 2001 Caffrey
6312412 November 6, 2001 Saied et al.
6317630 November 13, 2001 Gross et al.
6321070 November 20, 2001 Clark et al.
6321654 November 27, 2001 Robinson
6323780 November 27, 2001 Morris
6334070 December 25, 2001 Nova et al.
6364866 April 2, 2002 Furr et al.
6371939 April 16, 2002 Bergens et al.
6377848 April 23, 2002 Garde et al.
6387078 May 14, 2002 Gillespie, III
6398760 June 4, 2002 Danby
6405912 June 18, 2002 Giannou
6406455 June 18, 2002 Willis et al.
6411567 June 25, 2002 Niemiec et al.
6413236 July 2, 2002 Van Dyke
6425897 July 30, 2002 Overes et al.
6428517 August 6, 2002 Hochman et al.
6428528 August 6, 2002 Sadowski
6475181 November 5, 2002 Potter et al.
6478769 November 12, 2002 Parker
6478771 November 12, 2002 Lavi et al.
6494863 December 17, 2002 Shaw et al.
6500150 December 31, 2002 Gross et al.
6514230 February 4, 2003 Munk et al.
6529446 March 4, 2003 de la Huerga
6530900 March 11, 2003 Daily et al.
6530904 March 11, 2003 Edwards et al.
6535714 March 18, 2003 Melker et al.
6540672 April 1, 2003 Simonsen et al.
6540675 April 1, 2003 Aceti et al.
6544234 April 8, 2003 Gabriel
6551276 April 22, 2003 Mann et al.
6551298 April 22, 2003 Zhang
6554798 April 29, 2003 Mann et al.
6558320 May 6, 2003 Causey, III et al.
6560471 May 6, 2003 Heller
6565509 May 20, 2003 Say
6565533 May 20, 2003 Smith et al.
6569123 May 27, 2003 Alchas
6572584 June 3, 2003 Shaw et al.
6574166 June 3, 2003 Niemiec
6575939 June 10, 2003 Brunel
RE38189 July 15, 2003 Walker et al.
6585685 July 1, 2003 Staylor et al.
6585698 July 1, 2003 Packman et al.
6589158 July 8, 2003 Winkler
6595956 July 22, 2003 Gross et al.
6597794 July 22, 2003 Cole et al.
6599272 July 29, 2003 Hjertman et al.
6610271 August 26, 2003 Wermeling
6633796 October 14, 2003 Pool et al.
6641566 November 4, 2003 Douglas et al.
6645181 November 11, 2003 Lavi et al.
6648850 November 18, 2003 Landau
6659980 December 9, 2003 Moberg et al.
6676630 January 13, 2004 Landau et al.
6679862 January 20, 2004 Diaz et al.
6689093 February 10, 2004 Landau
6702778 March 9, 2004 Hill et al.
6706019 March 16, 2004 Parker et al.
6707763 March 16, 2004 Osberg et al.
6708050 March 16, 2004 Carim
6722916 April 20, 2004 Buccinna et al.
6723077 April 20, 2004 Pickup et al.
6726657 April 27, 2004 Dedig et al.
6726661 April 27, 2004 Munk et al.
6736796 May 18, 2004 Shekalim
6743635 June 1, 2004 Neel et al.
6749437 June 15, 2004 Chan
6752781 June 22, 2004 Landau et al.
6764469 July 20, 2004 Broselow
6767336 July 27, 2004 Kaplan
6770052 August 3, 2004 Hill et al.
6770056 August 3, 2004 Price et al.
6783509 August 31, 2004 Landau et al.
6784798 August 31, 2004 Morris
6786875 September 7, 2004 Barker et al.
6786885 September 7, 2004 Hochman et al.
6793646 September 21, 2004 Giambattista et al.
6803856 October 12, 2004 Murphy et al.
6805686 October 19, 2004 Fathallah et al.
6808514 October 26, 2004 Schneider et al.
6809653 October 26, 2004 Mann et al.
6817986 November 16, 2004 Slate et al.
6830560 December 14, 2004 Gross et al.
6839304 January 4, 2005 Niemiec et al.
6872200 March 29, 2005 Mann et al.
6875195 April 5, 2005 Choi
6883222 April 26, 2005 Landau
6923764 August 2, 2005 Aceti et al.
6936029 August 30, 2005 Mann et al.
6936032 August 30, 2005 Bush, Jr. et al.
6937150 August 30, 2005 Medema et al.
6942646 September 13, 2005 Langley et al.
6945961 September 20, 2005 Miller et al.
6946299 September 20, 2005 Neel et al.
6948492 September 27, 2005 Wermeling et al.
6949082 September 27, 2005 Langley et al.
6950028 September 27, 2005 Zweig
6952604 October 4, 2005 DeNuzzio et al.
6953445 October 11, 2005 Wilmot et al.
6953693 October 11, 2005 Neel et al.
6958691 October 25, 2005 Anderson et al.
6959247 October 25, 2005 Neel et al.
6961285 November 1, 2005 Niemiec et al.
6964650 November 15, 2005 Alexandre et al.
6969259 November 29, 2005 Pastrick et al.
6979316 December 27, 2005 Rubin et al.
6979326 December 27, 2005 Mann et al.
6985870 January 10, 2006 Martucci et al.
6997911 February 14, 2006 Klitmose
7003341 February 21, 2006 Say
7014470 March 21, 2006 Vann
7074211 July 11, 2006 Heiniger et al.
7093595 August 22, 2006 Nesbitt
7102526 September 5, 2006 Zweig
7104972 September 12, 2006 Moller et al.
7113101 September 26, 2006 Petersen et al.
7116233 October 3, 2006 Zhurin
7126879 October 24, 2006 Snyder
7158011 January 2, 2007 Brue
7158040 January 2, 2007 Morris
7190988 March 13, 2007 Say
7191916 March 20, 2007 Clifford et al.
7229458 June 12, 2007 Boecker et al.
7278983 October 9, 2007 Ireland et al.
7299981 November 27, 2007 Hickle et al.
7343914 March 18, 2008 Abrams et al.
7357790 April 15, 2008 Hommann et al.
7416540 August 26, 2008 Edwards et al.
7465294 December 16, 2008 Vladimirsky
7500963 March 10, 2009 Westbye et al.
7500967 March 10, 2009 Thorley et al.
7544188 June 9, 2009 Edwards et al.
7635348 December 22, 2009 Raven et al.
7648482 January 19, 2010 Edwards et al.
7648483 January 19, 2010 Edwards et al.
7654983 February 2, 2010 De La Sema et al.
7670328 March 2, 2010 Miller
7674246 March 9, 2010 Gillespie et al.
7682155 March 23, 2010 Raven et al.
7731686 June 8, 2010 Edwards et al.
7731690 June 8, 2010 Edwards et al.
7749194 July 6, 2010 Edwards et al.
7806866 October 5, 2010 Hommann et al.
7850662 December 14, 2010 Veasey et al.
7871393 January 18, 2011 Monroe
7901377 March 8, 2011 Harrison et al.
7910599 March 22, 2011 Sinclair
7918832 April 5, 2011 Veasey et al.
7938802 May 10, 2011 Bicknell et al.
7947017 May 24, 2011 Edwards et al.
8021335 September 20, 2011 Lesch, Jr.
8021344 September 20, 2011 Edwards et al.
8123719 February 28, 2012 Edwards et al.
8162886 April 24, 2012 Sadowski et al.
8198291 June 12, 2012 Wermeling
8206360 June 26, 2012 Edwards et al.
8226610 July 24, 2012 Edwards et al.
8231573 July 31, 2012 Edwards et al.
8544645 October 1, 2013 Edwards et al.
8627816 January 14, 2014 Edwards et al.
8647306 February 11, 2014 Schwirtz et al.
8690827 April 8, 2014 Edwards et al.
8734392 May 27, 2014 Stadelhofer
8734394 May 27, 2014 Adams et al.
8747357 June 10, 2014 Stamp et al.
20010005781 June 28, 2001 Bergens et al.
20020016567 February 7, 2002 Hochman et al.
20020072784 June 13, 2002 Sheppard, Jr. et al.
20020074345 June 20, 2002 Schneider et al.
20020076679 June 20, 2002 Aman
20020090601 July 11, 2002 Strupat et al.
20020095120 July 18, 2002 Larsen et al.
20020096543 July 25, 2002 Juselius
20020169439 November 14, 2002 Flaherty
20020183721 December 5, 2002 Santini, Jr. et al.
20030028145 February 6, 2003 Duchon et al.
20030040717 February 27, 2003 Saulenas et al.
20030060765 March 27, 2003 Campbell et al.
20030105430 June 5, 2003 Lavi et al.
20030106824 June 12, 2003 Wilmot et al.
20030120212 June 26, 2003 Dedig et al.
20030120222 June 26, 2003 Vaillancourt
20030135388 July 17, 2003 Martucci et al.
20030233070 December 18, 2003 De La Serna et al.
20040015125 January 22, 2004 Alexandre et al.
20040019326 January 29, 2004 Gilbert et al.
20040024367 February 5, 2004 Gilbert
20040039336 February 26, 2004 Amark et al.
20040039337 February 26, 2004 Letzing
20040039368 February 26, 2004 Reilly et al.
20040054327 March 18, 2004 Gillespie, III
20040078001 April 22, 2004 Langley et al.
20040116854 June 17, 2004 Abulhaj et al.
20040138611 July 15, 2004 Griffiths et al.
20040143298 July 22, 2004 Nova et al.
20040159364 August 19, 2004 Landau et al.
20040210199 October 21, 2004 Atterbury et al.
20040220524 November 4, 2004 Sadowski et al.
20040235731 November 25, 2004 Lundgren et al.
20040249358 December 9, 2004 McWethy et al.
20040267204 December 30, 2004 Brustowicz
20050027255 February 3, 2005 Lavi et al.
20050033234 February 10, 2005 Sadowski et al.
20050033386 February 10, 2005 Osborn et al.
20050038062 February 17, 2005 Burns et al.
20050055014 March 10, 2005 Coppeta et al.
20050062603 March 24, 2005 Fuerst et al.
20050088289 April 28, 2005 Rochkind
20050090781 April 28, 2005 Baba et al.
20050090782 April 28, 2005 Marshall et al.
20050092679 May 5, 2005 Warby
20050101912 May 12, 2005 Faust et al.
20050134433 June 23, 2005 Sweeney, II
20050137530 June 23, 2005 Campbell et al.
20050148931 July 7, 2005 Juhasz
20050148945 July 7, 2005 Chen
20050159705 July 21, 2005 Crawford et al.
20050165360 July 28, 2005 Stamp
20050168337 August 4, 2005 Mahoney
20050171477 August 4, 2005 Rubin et al.
20050177111 August 11, 2005 Ozeri et al.
20050182358 August 18, 2005 Veit et al.
20050183982 August 25, 2005 Giewercer
20050186221 August 25, 2005 Reynolds et al.
20050197654 September 8, 2005 Edman et al.
20050261742 November 24, 2005 Nova et al.
20050267403 December 1, 2005 Landau et al.
20050277891 December 15, 2005 Sibbitt
20060030819 February 9, 2006 Young et al.
20060053036 March 9, 2006 Coffman et al.
20060058848 March 16, 2006 Piraino et al.
20060083691 April 20, 2006 Wermeling
20060089592 April 27, 2006 Kadhiresan et al.
20060111666 May 25, 2006 Hommann et al.
20060111671 May 25, 2006 Klippenstein
20060116639 June 1, 2006 Russell
20060129090 June 15, 2006 Moberg et al.
20060189938 August 24, 2006 Hommann et al.
20060200077 September 7, 2006 Righi et al.
20060233778 October 19, 2006 Lundgren et al.
20060235354 October 19, 2006 Kaal et al.
20060247578 November 2, 2006 Arguendas et al.
20060247579 November 2, 2006 Friedman
20060265186 November 23, 2006 Holland et al.
20070008113 January 11, 2007 Spoonhower et al.
20070074722 April 5, 2007 Giroux et al.
20070100288 May 3, 2007 Bozeman et al.
20070111175 May 17, 2007 Raven et al.
20070129686 June 7, 2007 Daily et al.
20070135767 June 14, 2007 Gillespie, III et al.
20070148097 June 28, 2007 Finn et al.
20070149954 June 28, 2007 Hood et al.
20070166187 July 19, 2007 Song et al.
20070184847 August 9, 2007 Hansen et al.
20070185053 August 9, 2007 Linn
20070210147 September 13, 2007 Morrone et al.
20070212307 September 13, 2007 Wermeling et al.
20070213598 September 13, 2007 Howard et al.
20070233001 October 4, 2007 Burroughs et al.
20070239116 October 11, 2007 Follman et al.
20070239140 October 11, 2007 Chechelski et al.
20070260210 November 8, 2007 Conroy
20070261695 November 15, 2007 Kottayil et al.
20070265568 November 15, 2007 Tsals et al.
20070293826 December 20, 2007 Wall et al.
20080059133 March 6, 2008 Edwards et al.
20080111685 May 15, 2008 Olson et al.
20080171995 July 17, 2008 Vitullo et al.
20080188798 August 7, 2008 Weber
20080230057 September 25, 2008 Sutherland
20080255513 October 16, 2008 Kaal et al.
20080262443 October 23, 2008 Hommann et al.
20090093759 April 9, 2009 Judd et al.
20090143761 June 4, 2009 Cantor et al.
20090176834 July 9, 2009 Kottayil et al.
20090192486 July 30, 2009 Wilmot et al.
20090221962 September 3, 2009 Kaal et al.
20090240200 September 24, 2009 Heneveld et al.
20090318361 December 24, 2009 Noera et al.
20100010454 January 14, 2010 Marshall et al.
20100049125 February 25, 2010 James et al.
20100160894 June 24, 2010 Julian et al.
20100185148 July 22, 2010 Gillespie, III et al.
20100211005 August 19, 2010 Edwards et al.
20100212663 August 26, 2010 Vedrine et al.
20100286612 November 11, 2010 Cirillo
20100331354 December 30, 2010 Wermeling
20110092954 April 21, 2011 Jennings
20110098655 April 28, 2011 Jennings et al.
20110189259 August 4, 2011 Vasisht et al.
20120079718 April 5, 2012 Singer et al.
20120091026 April 19, 2012 Chacornac et al.
20120107783 May 3, 2012 Julian et al.
20120116319 May 10, 2012 Grunhut
20120226226 September 6, 2012 Edwards et al.
20120233834 September 20, 2012 Szechinski et al.
20120270895 October 25, 2012 Wermeling
20120302966 November 29, 2012 Vedrine
20130023822 January 24, 2013 Edwards et al.
20130023825 January 24, 2013 Edwards et al.
20130030367 January 31, 2013 Wotton et al.
20130060231 March 7, 2013 Adlon et al.
20130060232 March 7, 2013 Adlon et al.
20130079725 March 28, 2013 Shang
20130236872 September 12, 2013 Laurusonis et al.
20130266919 October 10, 2013 Baker et al.
20140052069 February 20, 2014 Edwards et al.
Foreign Patent Documents
0429039 March 1995 EP
0346830 May 1995 EP
1287840 March 2003 EP
1462134 September 2004 EP
1712178 October 2006 EP
1095668 April 2007 EP
PA04009276 January 2005 MX
WO 91/04760 April 1991 WO
WO 93/02720 February 1993 WO
WO 95/26009 September 1995 WO
WO 97/30742 August 1997 WO
WO 98/52632 November 1998 WO
WO 99/07425 February 1999 WO
WO 99/10031 March 1999 WO
WO 99/43283 September 1999 WO
WO 01/24690 April 2001 WO
WO 01/26020 April 2001 WO
WO 01/41849 June 2001 WO
WO 01/88828 November 2001 WO
WO 02/11778 February 2002 WO
WO 02/24257 March 2002 WO
WO 02/051471 July 2002 WO
WO 02/083205 October 2002 WO
WO 02/083212 October 2002 WO
WO 03/011378 February 2003 WO
WO 03/013632 February 2003 WO
WO 03/057283 July 2003 WO
WO 03/095001 November 2003 WO
WO 03/097133 November 2003 WO
WO 2004/041330 May 2004 WO
WO 2004/047890 June 2004 WO
WO 2004/047891 June 2004 WO
WO 2004/047892 June 2004 WO
WO 2004/047893 June 2004 WO
WO 2004/054644 July 2004 WO
WO 2005/020906 March 2005 WO
WO 2005/050526 June 2005 WO
WO 2005/070481 August 2005 WO
WO 2006/045525 May 2006 WO
WO 2005/077441 August 2006 WO
WO 2006/085175 August 2006 WO
WO 2006/085204 August 2006 WO
WO 2006/109778 October 2006 WO
WO 2006/125692 November 2006 WO
WO 2007/032962 March 2007 WO
WO 2007/088444 August 2007 WO
WO 2008/005315 January 2008 WO
WO 2008/148864 December 2008 WO
Other references
  • “Solutions for Medical Devices,” 3M Brochure, © 3M 2006 80-6201-3490-0.
  • Tingelstad, M., “Revolutionary Medical Technology Increases Demand for Flexible Interconnects,” [online] May 15, 2006 [retrieved on Nov. 15, 2006] Retrieved from the Internet <URL: http://www.ecnmag.com/index.asp?layout=articlePrint&ArticleID=CA6332947>.
  • “Flexible circuits / Flex circuits / Flexible Technology Ltd.,” Flexible Technology Limited [online] [retrieved on Aug. 28, 2006] Retrieved from the Internet <URL: http://www.flexibletechnology.com/>.
  • “Flexible circuits capabilities of Flexible Technology Limited,” Our Flexible Circuits Capabilities [online] [retrieved on Aug. 28, 2006] Retrieved from the Internet <URL: http://www.flexibletechnology.com/Flexible circuits Capability.htm >.
  • “Flex Circuits/flexible circuits design guide,” [online] [retrieved on Aug. 28, 2006] Retrieved from the Internet <URL: http://flexiblecircuit.co.uk/Flex Circuits Design Guide.htm >.
  • “Insect Stings Auto-injector Pouches and Carry Cases,” The Insect Stings On-Line Shop, [online] [retrieved on Jan. 24, 2007] Retrieved from the Internet <URL: http://www.insectstings.co.uk/acatalog/Auto Injector Pouches.html >.
  • “Anaphylaxis Canada Product Catalogue,” Anaphylaxis Canada > Living with Anaphylaxis > Tools and Resources [online] [retrieved on Jan. 24, 2007] Retrieved from the Internet <URL: http://anaphylaxis.om/content/livingwith/product catalogue.asp >.
  • “Microfluidics Device Provides Programmed, Long-Term Drug Dosing,” nano techwire.com [online] [retrieved on Nov. 28, 2006] Retrieved from the Internet <URL: http://nanotechwire.com/news.asp?nid=3141&ntid=124&pg=1 >.
  • Allan, R., “Medical Electronics: Technology Advances Will Revolutionize Healthcare,” Sep. 30, 2002 [online] [retrieved on Nov. 28, 2006] Retrieved from the Internet <URL: http://www.elecdesign.com/Articles/Index.cfm?AD=1&ArticleID=2041>.
  • RFID Gazette, “Smart Labels in Healthcare,” Sep. 29, 2005 [online] [retrieved on Nov. 28, 2006] Retrieved from the Internet <URL: http://www.rfidagazeete.org/2005/09/smart labels in.html >.
  • “Merck Serono Launches easypod(R), First Electronic Growth Hormone Injection Device,” Jan. 30, 2007 [online] [retrieved on Feb. 5, 2007] Retrieved from the Internet <URL: http://www.biz.yahoo.com/prnews/070130/ukm028.html?.v=8.
  • Scholz, O., “Drug depot in a tooth,” [online] [retrieved on Feb. 6, 2007] Retrieved from the Internet <URL: http://www.fraunhofer.de/fhg/EN/press/pi/2007/02Mediendienst22007Thema2.jsp?print=true.
  • Heartsine Technology, samaritan™ Pad Accessories [online] [retrieved on Jun. 1, 2007] Retrieved from the Internet <URL: http://www.heartsine.com/aboutsam-accessories.htm>.
  • CliniSense Corporation, “Drug delivery devices A potentially harsh environment for drugs,” Stability [online] [retrieved on Jun. 1, 2007] Retrieved from the Internet <URL: http://www.clinisense.com/devices.htm>.
  • CliniSense Corporation, “LifeTrack Technology a new method to detect improper storage.” Stability [online] [retrieved on Jun. 1, 2007] Retrieved from the Internet <URL: http://www.clinisense.com/tech.htm>.
  • AED Professionals™ Brochure [online] [retrieved on Jun. 1, 2007] Retrieved from the Internet <URL: http://www.aedprofessionals.com>.
  • Ruppar, D., “Implant Technologies Expected to Remain a Niche but Effective Method of Drug Delivery,” Drug Delivery Technology, Feb. 2007, vol. 7, No. 2 [online] [retrieved on Jun. 1, 2007] Retrieved from the Internet <URL: http://www.drugdeliverytech-online.com/drugdelivery/200702/templates/pageviewerprint?pg=44&pm=8 >.
  • O'Hagan, D. T. et al., “Novel approaches to pediatric vaccine delivery,” Advanced Drug Delivery Reviews, 58:29-51 (2006).
  • Boseley, S., “Families to receive antidote to help drug users who overdose,” Guardian News and Media, Jun. 25, 2009. [retrieved on May 27, 2011] Retrieved from the Internet <URL: http://www.guardian.co.uk/society/2009/jun/25/drug-verdose-antidote-naloxone-families>.
  • McDougall, L., “Addicts to be given personal supply of anti-overdose drug,” The Herald Scotland, May 28, 2006. Retrieved from the Internet <URL: http://www.heraldscotland.com/sport/spl/aberdeen/addicts-to-be-given-personal-supply-of-anti-overdose-drug-heroin-controversial-lifesaving-plan-projects-aim-to-cut-rising-death-toll-by-making-naloxone-treatment-more-readily-available-1.19181>.
  • BD Accuspray™ Nasal Spray System, 2004, Retrieved from the Internet <URL: http://www.bd.com/press/pdfs/flu/bdaccuspray.pdf>.
  • Colliver, V., “Naloxone saves lives of overdosed opiate users,” San Francisco Chronicle, Oct. 14, 2010 [retrieved on Aug. 14, 2013] Retrieved from the Internet <URL: http://www.sfgate.com/health/article/Naloxone-saves-lives-of-overdosed-opiate-users-3249978.php>.
  • Terry, D., “A Shot That Saves the Lives of Addicts Is Now in Their Hands,” The New York Times, Jul. 24, 2010 [retrieved on Aug. 14, 2013] Retrieved from the Internet <URL: http://www.nytimes.com/2010/07/25/us/25cncnaloxone.html>.
  • Szalavitz, M., “Should an Overdose Antidote Be Made More Accessible?” Time, Dec. 9, 2010 [retrieved on Aug. 14, 2013] Retrieved from the Internet <URL: http://healthland.time.com/2010/12/09/should-an-overdose-antidote-be-made-more-accessible/>.
  • Okie, S., “A Flood of Opioids, a Rising Tide of Deaths,” The New England Journal of Medicine, Nov. 18, 2010, 363:1981-1985 [online], [retrieved on Aug. 14, 2013] Retrieved from the Internet <URL: http://www.nejm.org/doi/full/10.1056/NEJMp1011512>.
  • Wermeling, D. P., “A response to the opioid overdose epidemic: naloxone nasal spray,” Drug Deliv. and Transl. Res., 3:63-74 (2013).
  • Djupesland, P. G., “Nasal drug delivery devices: characteristics and performance in a clinical perpsective—a review,” Drug Deliv. and Transl. Res., Published online: Oct. 18, 2012, 21 pages.
  • Examination Report for Australian Patent Application No. 2011218756, mailed Nov. 1, 2011.
  • Examination Report for Australian Patent Application No. 2011218756, mailed Feb. 1, 2013.
  • Notice of Acceptance for Australian Patent Application No. 2011218756, dated Apr. 11, 2013.
  • Non-Final Office Action for U.S. Appl. No. 13/036,720, mailed May 17, 2013.
  • Final Office Action for U.S. Appl. No. 13/036,720, mailed Nov. 5, 2013.
  • International Search Report and Written Opinion for International Application No. PCT/US2012/026708, mailed Jun. 7, 2012.
  • Wermeling, D. P., “Opioid Harm Reduction Strategies: Focus on Expanded Access to Intranasal Naloxone,” Pharmacotherapy, 30(7):627-631 (2010).
  • “Martindale Pharma launches Prenoxad Injection in UK for Emergency Treatment of Opioid Overdose,” Pharmabiz.com, May 6, 2013 [online], [retrieved on Nov. 26, 2014] Retrieved from the Internet <URL: http://www.pharmabiz.com/NewsDetails.aspx?aid=75187&sid=2>, 1 page.
  • “Therapeutic Intranasal Drug Delivery: Needleless Treatment Options for Medical Problems,” Intranasal.net [online], [retrieved on Dec. 16, 2010] Retrieved from the Internet <URL: http://intranasal.net/OpiateOverdose/>, 10 pages.
  • Prenoxad Injection: Client's Guide to Prenoxad Injection, Martindale Pharma, Apr. 2013, 16 pages.
  • Prenoxad Injection: Pharmacists's Guide, Martindale Pharma, Apr. 2013, 6 pages.
  • Prenoxad Injection: Packaging Leaflet: Information for the User, Martindale Pharma, Nov. 2012, 2 pages.
  • Prenoxad Injection: Summary of Product Characteristics, Martindale Pharma, Apr. 2013, 4 pages.
  • Edwards, E. S., Dissertation: “Development of a novel approach to assess qualitative and quantitative dynamics associated with the subcutaneous or intramuscular administration of pharmaceuticals and associated parenteral delivery systems,” B.S. Biology, Virginia Commonwealth University (Dec. 2011), 316 pages.
  • Bennett, J. et al., “Subcutaneous administration of midazolam: A comparison of the bioject jet injector with the conventional syringe and needle,” J Oral Maxillofac Surg, 56(11):1249-1254 (1998).
  • Brearley, C. et al., “Pharmacokinetics of recombinant human growth hormone administered by cool.clickTM 2, a new needle-free device, compared with subcutaneous administration using a conventional syringe and needle,” BMC Clin Pharrnacol, 7:10 (2007), 7 pages.
  • Simons, F. E. R. et al., “Epinephrine absorption in adults: intramuscular versus subcutaneous injection,” J Allergy Clin Immunol, 108(5), 871-873 (2001).
  • Kerum, G. et al., “Blood glucose and free insulin levels after the administration of insulin by conventional syringe or jet injector in insulin treated type 2 diabetics,” Horm. Metabol. Res., 19:422-425 (1987).
  • Halle, J.-P. et al., “Twice daily mixed regular and NPH insulin injections with new jet injector versus conventional syringes: pharmacokinetics of insulin absorption,” Diabetes Care, 9(3):279-282 (1986).
  • Taylor, R. et al., “Plasma free insulin profiles after administration of insulin by jet and conventional syringe injection,” Diabetes Care, 4(3):377-379 (1981).
  • Biocryst Pharmaceuticals, Inc., Biocryst Press Release, “Biocryst reports preliminary results from a phase II clinical trial of peramivir in subjects with acute influenza,” Retrieved from the Internet: <http://investor.shareholder.com/biocryst/releasedetail.cfm?ReleaseID=264815> (Sep. 19, 2007), 3 pages.
Patent History
Patent number: 9022022
Type: Grant
Filed: Jan 13, 2014
Date of Patent: May 5, 2015
Patent Publication Number: 20140123974
Assignee: kaleo, Inc. (Richmond, VA)
Inventors: Eric S. Edwards (Moseley, VA), Evan T. Edwards (Charlottesville, VA), Mark J. Licata (Doswell, VA), Frank E. Blondino (Henrico, VA)
Primary Examiner: Kami A Bosworth
Assistant Examiner: Imani Hayman
Application Number: 14/153,575
Classifications
Current U.S. Class: Liquid Medicament Atomizer Or Sprayer (128/200.14); Treating Material Forced Into Or Out Of Body By Self-acting Fluid Pressure, Motor-driven, Or Mechanical Energy Storing Means (e.g., Pressure Infusion Or Aspiration, Etc.) (604/131)
International Classification: A61M 11/00 (20060101); A61M 37/00 (20060101); A61M 5/20 (20060101); A61M 5/32 (20060101); A61M 15/08 (20060101);